WO2008048648A2 - Cb1-modulating compounds and their use - Google Patents

Cb1-modulating compounds and their use Download PDF

Info

Publication number
WO2008048648A2
WO2008048648A2 PCT/US2007/022183 US2007022183W WO2008048648A2 WO 2008048648 A2 WO2008048648 A2 WO 2008048648A2 US 2007022183 W US2007022183 W US 2007022183W WO 2008048648 A2 WO2008048648 A2 WO 2008048648A2
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
compound
alkyl
group
heteroalicyclyl
Prior art date
Application number
PCT/US2007/022183
Other languages
French (fr)
Other versions
WO2008048648A3 (en
Inventor
Roger Olsson
Anne Bulow
Original Assignee
Acadia Pharmaceuticals 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 Acadia Pharmaceuticals Inc. filed Critical Acadia Pharmaceuticals Inc.
Publication of WO2008048648A2 publication Critical patent/WO2008048648A2/en
Publication of WO2008048648A3 publication Critical patent/WO2008048648A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-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
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence

Definitions

  • This invention relates to the fields of organic chemistry, pharmaceutical chemistry, biochemistry, molecular biology and medicine.
  • it relates to compounds that modulate the activity of the human cannabinoid receptor (CBl), and to the use of the compounds for the treatment and prevention of diseases and disorders related to CB 1.
  • CBl cannabinoid receptor
  • the cannabinoids which are bioactive lipids, naturally found in the cannabis sativa (marijuana) plant, have been used recreationally and therapeutically for at least 5000 years. In addition to their well-documented effects on mood, cannabinoids (often in the form of marijuana) have been prescribed to treat nausea, pain, migraine, epilepsy, glaucoma, hypertension, cachexia and pain associated with childbirth.
  • Two cannabinoid receptors, CBl and CB2 have been identified. Both are members of the G protein-coupled receptor superfamily, and are negatively coupled through Gi protein. The CB2 receptor has 44% sequence similarity to the CBl receptor.
  • the CB 1 receptor unlike the CB2 receptor, is highly expressed in the central nervous system, mostly presynaptically. Indeed, the CBl receptor is present in the brain at higher levels than many other GPCRs. It is found in the cortex, cerebellum, hippocampus, and basal ganglia (reviewed in Brievogel and Childres, 1998). In addition, the CBl receptor has also been detected in sperm, the prostate gland, and other peripheral tissues (including structures of the eye). The CB2 receptor is present in the cells of the immune system (spleen, thymus), testis, and lung.
  • the CBl receptor is believed to be responsible for the appetite stimulating properties and habituation associated with cannabinoid use.
  • the CBl receptor antagonist, SR141716 (rimonabant, Acomplia; Sanof ⁇ -Aventis) has shown efficacy in late-stage clinical trials for obesity and nicotine dependence, with no psychotropic effects. The compound has been shown to reduce both food intake and adipose tissue (by a mechanism independent of food intake).
  • Use of SR141716 in animal models suggests additional use of CB 1 receptor antagonists and inverse agonists for the treatment of alcohol addiction, opiate addiction, ***e addiction, anxiety, and septic shock.
  • mice null for the CBl gene also display impaired ***e self- administration, and less severe withdrawal from morphine addiction compared to wild- type mice.
  • CBl knockout mice also display increased bone mineral density, and both CBl knockout mice and mice treated with CB antagonists are resistant to bone loss in a model for osteoporosis.
  • Other animal models indicate a use for CBl receptor antagonists and inverse agonists for the prevention of premature spontaneous abortion.
  • Cannabinoid signaling is hyperactive in animal models of several diseases suggesting that cannabinoids either have a protective role (e.g., CBl agonists may be therapeutic) or are involved in the pathology of these diseases (e.g., CBl antagonists or inverse agonists may be therapeutic). These include Parkinson's disease, Alzheimer's disease, multiple sclerosis, epilepsy, and intestinal disorders. In addition, the levels of endogenous cannabinoids and CBl receptors are elevated in the liver and blood of patients with cirrhosis of the liver. Moreover, cannabinoid levels have been shown to be elevated in the cerebrospinal fluid of a patient with stroke, as well as in the brains of depressed suicide victims.
  • Endogenous cannabinoids have also been shown to be higher in the cerebrospinal fluid of drug-naive paranoid schizophrenics compared to normal patients; interestingly, schizophrenic patients treated with atypical but not typical antipsychotics also exhibit higher CSF levels of anandamide. Additionally, the CBl gene is located in a chromosomal region that has been linked to schizophrenia. Moreover, high levels of the endogenous cannabinoid, anandamide, are correlated with premature abortion and failure of in vitro fertilization. Finally, activation of CB receptors by an anandamide analogue has been shown to reduce sperm fertilizing capacity by 50%.
  • CBl receptors by agonists or partial agonists may also be used to treat a number of disorders.
  • THC tetrahydrocannabinol; active cannabinoid in Cannabis sativa
  • cannabinoids have been shown to improve mobility and alleviate pain in patients with multiple sclerosis.
  • cannabinoids have been shown in clinical trials for Tourette's syndrome, Parkinson's disease, glaucoma, and pain.
  • cannabinoids have been shown to inhibit cancer growth, angiogenesis, and metastasis in animal models.
  • compositions comprising a compound of Formula (I).
  • a method of treating a disease and/or condition that would be alleviated, ameliorated, and/or treated by administration of a compound that modulates a cannabinoid receptor comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
  • D can be part of Ar 1 and is selected from CRi, NR 2 , S, and O.
  • R 2 can be absent or is selected from hydrogen; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, and alkynyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl.
  • R 3 , R 3a , and R 3b can be each independently selected from: hydrogen; mono-substituted, poly- substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, aryl.lieteroaryl, and heteroalicyclyl.
  • Z can be O or S.
  • B is not selected from -CF 3 , phenyl, -OS(O) 2 -
  • B is not halogen when A is substituted or unsubstituted aryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroaryl, halogen, or substituted or unsubstituted sulfenyl;
  • B when X is CR ⁇ Ri b and A is phenyl, then B cannot be NH 2 .
  • X when both Ari and Ar 2 are pyridinyl rings, then X cannot be NRj in which Ri is selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, trihalomethyl and hydroxyalkyl.
  • Ri when X is S or NRi, wherein Ri is hydrogen or alkyl, then A cannot be a phenyl ring substituted at the para-position with -CO 2 H or CO 2 (alkyl).
  • Ri 3 and Ri b can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms and substituted with subtituents selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido
  • R 4 and R 5 are each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxyl, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyana
  • Rib can be hydrogen. In other embodiments,
  • Ri b can be Ci -3 alkyl.
  • X can be S (sulfur).
  • B can be selected from cycloalkyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted.
  • B can be an unsubstituted or substituted heteroaryl selected from:
  • B can be an optionally substituted phenyl.
  • the optionally substituted phenyl can be substituted with a C M alkyl.
  • Ria can be selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH 2 ) 0-7 -NR 3a R 3b , wherein any member of said group can be substituted or unsubstituted.
  • Ri a can be selected from hydrogen, alkyl, alkoxy, aryl, aralkyl, heteroaryl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted. In some embodiments, Ri a can be an optionally substituted heteroaryl or heteroaralkyl.
  • heteroaralkyl can be selected from: , wherein Q is oxygen or sulfur, and in some embodiments, n can be 1 or 2. hi more particular embodiments, the optionally substituted heteroaralkyl can be
  • Rn can be hydrogen.
  • B can be -S(O)NRi a Ri b or -S(O) 2 NRi a Ri b .
  • B can be -S(O)NRi a Rib or -S(O) 2 NRi 3 RIb and Ri a can be selected from alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl.
  • B can be - S(O) 2 NRi a Ri b and Ri a can be selected from alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl.
  • B can be -S(O) 2 NRi a Ri b and Ri a can be alkyl or heteroaryl.
  • R ⁇ can be hydrogen.
  • R )a and Ri b can each independently selected from:
  • n can be an integer selected from O, 1, 2, 3, 4, 5, 6 or 7 defining the number of optionally substituted carbon atoms;
  • Q can be selected from -N(R 4 )-, O and S;
  • R 4 and R 5 each each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato,
  • R 6 , R ⁇ a, R ⁇ b, R ⁇ c, and R 61 I can each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S- sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyana
  • R ⁇ can be hydrogen.
  • a , R 2a , R 2 , R 3 , R 3a , and R 3b can be each independently selected from aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, or (heteroalicyclyl)alkyl and are substituted with zero to five substituents, wherein each substituent is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, .
  • Ri ; R la and R ⁇ can be independently selected from f hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl.
  • Ri and Ri 3 can be independently selected from of alkyl, cycloalkyl, heteroaryl, heteroalicyclyl and heteroaralkyl; and R ⁇ can be hydrogen.
  • A can be an aryl, heteroaryl, or heteroalicyclyl, and is substituted with zero to five substituents, wherein each substituent is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-car
  • A can be an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent can be independently selected from alkyl, alkoxy, ester, cyano, and halogen.
  • the heteroaryl can be substituted or unsubstituted thiophene or substituted or unsubstituted pyridine.
  • the aryl can be an unsubstituted or substituted phenyl (e.g., 2-, 3-, 4-, 2-,3-, 2-,4- substituted phenyl).
  • A can be an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent is independently selected from alkyl, alkoxy, ester, cyano, and halogen.
  • the phenyl when A is substituted phenyl, the phenyl can be substituted with a halogen, methoxy, or cyano group.
  • A can be alkyl or aryl. In other embodiments, A can be cycloalkyl.
  • A can be selected from C 3 -Ci 2 alkyl, C 4 - Ci 2 alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstiruted;
  • B, C, D, E, F, G and I can be separately selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aral
  • R 2 can be absent or is selected from: hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstitute
  • Z can be O (oxygen).
  • A can be selected from C 3 - Ci 2 alkyl (e.g., n-propyl), C 4 -Ci 2 alkyl (e.g., n-butyl), cycloalkyl (e.g., cyclohexyl), aryl (e.g., substituted or unsubstituted phenyl), and heteroaryl (e.g., thiophene and pyridine), wherein any member of said group can be substituted or unsubstituted.
  • Z can be O (oxygen) and A can be selected from C 3 -Ci 2 alkyl (e.g., n- propyl), C4-Ci 2 alkyl (e.g., n-butyl), cycloalkyl (e.g., cyclohexyl), aryl (e.g., substituted or unsubstituted phenyl), and heteroaryl (e.g., thiophene and pyridine), wherein any member of said group can be substituted or unsubstituted.
  • A can be selected from C 3 -Ci 2 alkyl (e.g., n- propyl), C4-Ci 2 alkyl (e.g., n-butyl), cycloalkyl (e.g., cyclohexyl), aryl (e.g., substituted or unsubstituted phenyl), and heteroaryl (e.g., thiophene and pyr
  • C 3 -Ci 2 alkyl e.g., n- propyl
  • C 4 -Ci 2 alkyl e.g., n-butyl
  • C 3 -Ci 2 alkyl e.g., n-propyl
  • C 4 -Ci 2 alkyl e.g., n-butyl
  • A can be -NR ⁇ Ri b wherein Ri 3 is an aryl (e.g., optionally substituted phenyl) and R ⁇ is hydrogen.
  • A can be -NRuRib wherein Ri a is a phenyl group substituted with a halogen and R ⁇ is hydrogen.
  • A can be C 3 -Ci 2 alkyl (e.g., n-propyl), C 4 -Ci 2 alkyl (e.g., n- butyl).
  • A can be cycloalkyl (e.g., cyclohexyl).
  • A can be aryl (e.g., substituted or unsubstituted phenyl).
  • the aryl can be an unsubstituted or substituted phenyl (e.g., 2-, 3-, 4-, 2-,3-, 2-,4- substituted phenyl)
  • A can be heteroaryl (e.g., optionally thiophene or optionally substituted pyridine).
  • A is not C 3 -, C 4 -, C 5 -, C 6 -, C 7 -, C 8 -, C 9 -, Ci 0 -, Cn-, Ci 2 alkyl.
  • X can be S (sulfur); and
  • X can be S;
  • X can be S;
  • A can be an aryl or a heteroaryl group;
  • A can be a cycloalkyl, a heteroalicyclyl, or - NRi a Ri b group;
  • X can be S;
  • X can be S;
  • A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRuRib group;
  • X can be S;
  • the alkyl can be Ci -6 alkyl.
  • the alkoxy is a Ci -6 alkoxy.
  • X can be S;
  • X can be S;
  • B can be wherein Ri 2 is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl.
  • A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or - NRi a Ri b group;
  • X can be S;
  • the optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl is selected from: of the embodiments, n can be 1 or 2.
  • X can be S;
  • X can be S;
  • A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • Y can be -N(R 2 ) ⁇ wherein the symbol ⁇ represents a double bond and R 2 does not exist;
  • A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRi a Ri b group;
  • X can be S;
  • substituted aryl or aralkyl can be selected from: ,wherein Q can be -N(R 4 )-, oxygen or sulfur; and R 4 can be hydrogen or Chalky!, and in some of the embodiments, n can be 1 or 2.
  • A can be selected from C 3 -Ci 2 alkyl, C 4 - Ci 2 alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NR ⁇ Ri b , and -(CH 2 ) 0 .
  • X can be S
  • X can be S;
  • A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or - NRi a Ri b group;
  • X can be S;
  • the optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl can be selected from:
  • X can be S;
  • X can be S;
  • A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRi a Ri b group;
  • X can be S;
  • the optionally substituted heteroaralkyl is from the
  • n 1 or 2.
  • the optionally substituted heteroaralkyl can ,wherein Q can be oxygen or sulfur, and in some of the embodiments, n can be 1 or 2.
  • X can be S; and A can be selected from a mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl and alkynyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl and heteroaralkyl.
  • A can be selected from a mono-substituted, poly- substituted, or unsubstituted, straight or branched alkyl; and mono-substituted, poly- substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl.
  • A can be a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; or mono-substituted, poly-substituted or unsubstituted aryl.
  • A can be a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; or mono-substituted, poly-substituted or unsubstituted aryl;
  • B can be -S(O)2NRi a Rib-
  • Z can be O (oxygen).
  • Ri 1 Ri 3 and Ri b can be independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl.
  • Ri and Ri a can be independently selected from alkyl, cycloalkyl, heteroaryl, heteroalicyclyl and heteroaralkyl; and R ⁇ is hydrogen.
  • Ari is phenyl.
  • Ari is selected from optionally substituted pyrazine, optionally substituted pyridine, optionally substituted pyrimidine, optionally substituted oxazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted pyrrole, optionally substituted imidazole, and optionally substituted phenyl.
  • Ar 2 is selected from optionally substituted pyrazine, optionally substituted pyridine, optionally substituted indole, optionally substituted pyrimidine, optionally substituted oxazole, optionally substituted thiazole, optionally substituted furan, optionally substituted thiophene, optionally substituted pyrrole, optionally substituted imidazole, optionally substituted triazaole, optionally substituted isoxazole, optionally substituted isothiazole, optionally substituted pyrazole, and optionally substituted phenyl.
  • the compound of Formula (I) can further include a detectable label such as a radiolabel.
  • detectable labels include, but are not limited to [ 3 H], [ 18 F], [ 11 C] and [ 125 I].
  • compounds according to Formula (I) include the following structures:
  • Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of X can be combined with any one or more embodiments of Y, A, Ar 1 , Ar 2 , D, B, R 1 , R 1 ,, R, b , R 2 , R 3 , R-3a, and R 3b .
  • Still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ari can be combined with any one or more embodiments of X, Y, A, Ar 2 , D, B, R 1 , R la , Rib, R2, R3, R3a, and R 3b .
  • Yet still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ar 2 can be combined with any one or more embodiments of X, Y, A, An, D, B, Ri, Ri 3 , R n ,, R 2 , R 3 , R 3a and R 3b .
  • Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of D can be combined with any one or more embodiments of X, Y, A, An, Ar 2 , B, R 1 , R la , R n ,, R 2 , R 3 , R-3a and R 3b .
  • Still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ri can be combined with any one or more embodiments of X, Y, A, Ari, Ar 2 , B, D, Ri a , Rn,, R 2 , R3, R 3 a and R 3b .
  • any embodiment of R) a can be combined with any one or more embodiments of X, Y, A, Ari, Ar 2 , B, D, Ri, Rn,, R 2 , R 3 , R 3a and R 3b .
  • Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ri b can be combined with any one or more embodiments of X, Y, A, Ar,, Ar 2 , B, D, R 1 , R la , R 2 , R 3 , R 3a and R 3b .
  • Still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R 3 can be combined with any one or more embodiments of X, Y, A, Ar 1 , Ar 2 , B, D, R t , Ri 3 , Ri b , R 2 , R 3a and R 3b .
  • any embodiment of R 3a can be combined with any one or more embodiments of X, Y, A, An, Ar 2 , B, D, Ri, Ri a , Ri b , R 2 , R 3 and R 3b .
  • Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R 3b can be combined with any one or more embodiments of X, Y, A, Ar 1 , Ar 2 , B, D, R 1 , R, a , R, b , R 2 , R 3 and R 3a .
  • Certain of the compounds of the present invention may exist as stereoisomers including optical isomers. The invention includes all stereoisomers and both the racemic mixtures of such stereoisomers as well as the individual enantiomers that may be separated according to methods that are well known to those of ordinary skill in the art.
  • the compound of Formula (I) can bind to a cannabinoid receptor.
  • the cannabinoid receptor can be a CBl receptor.
  • any "R" group(s) such as, without limitation, Ri, R !a and Ri b , represent substituents that can be attached to the indicated atom.
  • R groups include but are not limited to hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl.
  • An R group may be substituted or unsubstituted.
  • R a and R b of an NR 3 R b group are indicated to be “taken together", it means that they are covalently bonded to one another at their terminal atoms to form a ring that includes the nitrogen:
  • IC 50 refers to an amount, concentration, or dosage of a particular test compound that achieves a 50% inhibition of a maximal response, such as modulation of GPCR, including cannabinoid receptor, activity an assay that measures such response.
  • the assay may be an R-SAT ® assay as described herein but is not limited to an RSAT assay.
  • EC 50 refers to an amount, concentration or dosage of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound, in an assay that measures such response such as but not limited to R-SAT ® assay described herein.
  • substituent is a group that may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N
  • C m to C n in which "m” and “n” are integers refers to the number of carbon atoms in an alkyl, alkenyl or alkynyl group or the number of carbon atoms in the ring of a cycloalkyl or cycloalkenyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl or ring of the cycloalkenyl can contain from “m” to "n", inclusive, carbon atoms.
  • a "Ci to C 4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH 3 -, CH 3 CH 2 -, CH 3 CH 2 CH 2 -, (CH 3 ) 2 CH-, CH 3 CH 2 CH 2 CH 2 -, CH 3 CH 2 CH(CH 3 )- and (CH 3 ) 3 C-. If no "m” and "n” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl or cycloalkenyl group, the broadest range described in these definitions is to be assumed.
  • alkyl refers to a straight or branched hydrocarbon chain fully saturated (no double or triple bonds) hydrocarbon group.
  • the alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., "1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term "alkyl” where no numerical range is designated).
  • the alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms.
  • the alkyl group could also be a lower alkyl having 1 to 5 carbon atoms.
  • the alkyl group of the compounds may be designated as "CpC 4 alkyl” or similar designations.
  • Ci-C 4 alkyl indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, and the like.
  • the alkyl group may be substituted or unsubstituted.
  • the substituent group(s) is(are) one or more group(s) individually and independently selected from alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-
  • alkenyl refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds.
  • An alkenyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
  • alkynyl refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds.
  • An alkynyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
  • aryl refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system that has a fully delocalized pi-electron system throughout all the rings.
  • aryl groups include, but are not limited to, benzene, naphthalene and azulene.
  • An aryl group of this invention may be substituted or unsubstituted.
  • substituent group(s) that is(are) one or more group(s) independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, iso
  • heteroaryl refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system throughout all the rings), one or two or more fused rings that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur.
  • heteroaryl rings include, but are not limited to, furan, thiophene, phthalazine, pyrrole, oxazole, thiazole, imidazole, pyrazole, isoxazole, isothiazole, triazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine and triazine.
  • a heteroaryl group of this invention may be substituted or unsubstituted.
  • substituent group(s) that is(are) one or more group(s) independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, iso
  • an "aralkyl” is an aryl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, substituted benzyl, 2- phenylethyl, 3-phenylpropyl, and naphtylalkyl.
  • a “heteroaralkyl” is heteroaryl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylmethyl, 3- thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl, and their substituted as well as benzo-fused analogs.
  • Lower alkylene groups are straight-chained tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (- CH 2 CH 2 CH 2 -), and butylene (-(CH 2 ) 4 -) groups. A lower alkylene group may be substituted or unsubstituted.
  • arylalkylidene refers to an alkylidene group in which either R' and R" is an aryl group. An alkylidene group may be substituted or unsubstituted.
  • alkoxy refers to the formula -OR wherein R is an alkyl is defined as above, e.g. methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, amoxy, tert-amoxy and the like.
  • An alkoxy may be substituted or unsubstituted.
  • alkylthio refers to the formula -SR wherein R is an alkyl is defined as above, e.g. methylmercapto, ethylmercapto, n-propylmercapto, 1- methylethylmercapto (isopropylmercapto), n-butylmercapto, iso-butylmercapto, sec- butylmercapto, tert-butylmercapto, and the like.
  • An alkylthio may be substituted or unsubstituted.
  • aryloxy and arylthio refers to RO- and RS-, in which R is an aryl, such as but not limited to phenyl. Both an aryloxy and arylthio may be substituted or unsubstituted.
  • acyl refers to a hydrogen, alkyl, alkenyl, alkynyl, or aryl connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl, and acryl. An acyl may be substituted or unsubstituted. An acyl may be substituted or unsubstituted.
  • cycloalkyl refers to a completely saturated (no double bonds) mono- or multi- cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro-connected fashion. Cycloalkyl groups of this invention may range from C 3 to Ci 0 , in other embodiments it may range from C 3 to C 6 . A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. If substituted, the substituent(s) may be an alkyl or selected from those indicated above with regard to substitution of an alkyl group unless otherwise indicated.
  • cycloalkenyl refers to a cycloalkyl group that contains one or more double bonds in the ring although, if there is more than one, they cannot form a fully delocalized pi-electron system in the ring (otherwise the group would be "aryl,” as defined herein). When composed of two or more rings, the rings may be connected together in a fused, bridged or spiro-connected fashion.
  • a cycloalkenyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be an alkyl or selected from the groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
  • cycloalkynyl refers to a cycloalkyl group that contains one or more triple bonds in the ring. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro-connected fashion.
  • a cycloalkynyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be an alkyl or selected from the groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
  • heteroalicyclic or “heteroalicyclyl” refers to a stable 3- to 18 membered ring which consists of carbon atoms and from one to five heteroatoms selected from nitrogen, oxygen and sulfur.
  • the "heteroalicyclic” or “heteroalicyclyl” may be monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may be joined together in a fused, bridged or spiro-connected fashion; and the nitrogen, carbon and sulfur atoms in the "heteroalicyclic” or “heteroalicyclyl” may be optionally oxidized; the nitrogen may be optionally quatemized; and the rings may also contain one or more double bonds provided that they do not form a fully delocalized pi-electron system throughout all the rings.
  • Heteroalicyclyl groups of this invention may be unsubstituted or substituted.
  • the substituent(s) may be one or more groups independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, iso
  • heteroalicyclic or “heteroalicyclyl” include but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, mo ⁇ holinyl, oxiranyl, piperidinyl N-Oxide, piperidinyl, piperazinyl, pyrrolidinyl, 4-piperidonyl, pyrazolidinyl, 2-oxopyrrolidinyl, thiamo ⁇ holinyl, thiamorpholinyl sulfoxide, and thiamo ⁇ holinyl sulfone unless the substituent groups are otherwise indicated.
  • a "(heteroalicyclyl)alkyl” is a heterocyclic or a heterocyclyl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and heterocyclic or a heterocyclyl of a (heteroalicyclyl)alkyl may be substituted or unsubstituted. Examples include but are not limited 4-methyltetrahydro-2H-pyran, substituted 4-methyltetrahydro-2H-pyran, 4-ethylpiperidine, 4-propylpiperidine, 4- methyltetrahydro-2H-thiopyran, and 4-methyl-l,3-thiazinane.
  • a "(cycloalkyl)alkyl” is a cycloalkyl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and cycloalkyl of a (cycloalkyl)alkyl may be substituted or unsubstituted.
  • Examples include but are not limited cyclopropylmethyl, cyclobutylmethyl, cyclopropylethyl, cyclopropylbutyl, cyclobutylethyl, cyclopropylisopropyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, cycloheptylmethyl, and the like.
  • a "(cycloalkenyl)alkyl” is a cycloalkenyl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and cycloalkenyl of a (cycloalkenyl)alkyl may be substituted or unsubstituted.
  • a "(cycloalkynyl)alkyl” is a cycloalkynyl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and cycloalkynyl of a (cycloalkynyl)alkyl may be substituted or unsubstituted.
  • halo or “halogen” refers to F (fluoro), Cl (chloro), Br (bromo) or I (iodo).
  • haloalkyl refers to an alkyl group in which one or more of the hydrogen atoms are replaced by halogen. Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifiuoromethyl and l-chloro-2- fluoromethyl, 2-fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.
  • haloalkoxy refers to RO-group in which R is a haloalkyl group.
  • groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy and l-chloro-2-fluoromethoxy, 2- fluoroisobutyoxy.
  • a haloalkoxy may be substituted or unsubstituted.
  • R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein.
  • An O- carboxy may be substituted or unsubstituted.
  • C-carboxy refers to a "-C(O)R” group in which R can be the same as defined with respect to O-carboxy.
  • a C-carboxy may be substituted or unsubstituted.
  • a "trihalomethanesulfonyl” group refers to an "X 3 CSO 2 -" group wherein X is a halogen.
  • a "cyano" group refers to a "-CN” group.
  • An "isocyanato” group refers to a "-NCO” group.
  • a "thiocyanato" group refers to a "-CNS” group.
  • An "isothiocyanato" group refers to an " -NCS” group.
  • a “sulfinyl” group refers to an "-S(O)-R" group in which R can be the same as defined with respect to O-carboxy.
  • a sulfinyl may be substituted or unsubstituted.
  • a “sulfonyl” group refers to an “SO 2 R” group in which R can be the same as defined with respect to O-carboxy.
  • a sulfonyl may be substituted or unsubstituted.
  • S-sulfonamido refers to a "-SO 2 NR A R B " group in which R A and R B can be the same as R defined with respect to O-carboxy.
  • An S-sulfonamido may be substituted or unsubstituted.
  • An "N-sulfonamido” group refers to a "RSO 2 N(R A )-” group in which R and R A can be the same as R defined with respect to O-carboxy.
  • a sulfonyl may be substituted or unsubstituted.
  • a "trihalomethanesulfonamido" group refers to an "X 3 CSO 2 N(R)-" group with X as halogen and R can be the same as defined with respect to O-carboxy.
  • a trihalomethanesulfonamido may be substituted or unsubstituted.
  • An O-carbamyl may be substituted or unsubstituted.
  • An N-carbamyl may be substituted or unsubstituted.
  • An O-thiocarbamyl may be substituted or unsubstituted.
  • An N-thiocarbamyl may be substituted or unsubstituted.
  • a C-amido may be substituted or unsubstituted.
  • An N-amido may be substituted or unsubstituted.
  • An ester may be substituted or unsubstituted.
  • a lower aminoalkyl refers to an amino group connected via a lower alkylene group.
  • a lower aminoalkyl may be substituted or unsubstituted.
  • a lower alkoxyalkyl refers to an alkoxy group connected via a lower alkylene group.
  • a lower alkoxyalkyl may be substituted or unsubstituted.
  • Any unsubstituted or monosubstituted amine group on a compound herein can be converted to an amide, any hydroxyl group can be converted to an ester and any carboxyl group can be converted to either an amide or ester using techniques well- known to those skilled in the art (see, for example, Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley & Sons, New York, NY, 1999).
  • haloalkyl may include one or more of the same or different halogens.
  • Ci-C 3 alkoxyphenyl may include one or more of the same or different alkoxygroups containing one, two or three atoms.
  • each center may independently be of R-configuration or S -configuration or a mixture thereof.
  • the compounds provided herein may be enatiomerically pure or be stereoisomeric mixtures.
  • each double bond may independently be E or Z a mixture thereof.
  • all tautomeric forms are also intended to be included.
  • salts refers to a salt of a compound that does not abrogate the biological activity and properties of the compound.
  • Pharmaceutical salts can be obtained by reaction of a compound disclosed herein with an acid or base.
  • Base-formed salts include, without limitation, ammonium salt (NH 4 + ); alkali metal, such as, without limitation, sodium or potassium, salts; alkaline earth, such as, without limitation, calcium or magnesium, salts; salts of organic bases such as, without limitation, dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine; and salts with the amino group of amino acids such as, without limitation, arginine and lysine.
  • NH 4 + ammonium salt
  • alkali metal such as, without limitation, sodium or potassium
  • alkaline earth such as, without limitation, calcium or magnesium
  • salts of organic bases such as, without limitation, dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine
  • Useful acid-based salts include, without limitation, hydrochlorides, hydrobromides, sulfates, nitrates, phosphates, methanesulfonates, ethanesulfonates, p-toluenesulfonates and salicylates.
  • solvates and hydrates are complexes of a compound with one or more solvent of water molecules, or 1 to about 100, or 1 to about 10, or one to about 2, 3 or 4, solvent or water molecules.
  • a "prodrug” refers to a compound that may not be pharmaceutically active but that is converted into an active drug upon in vivo administration.
  • the prodrug may be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
  • Prodrugs are often useful because they may be easier to administer than the parent drug. They may, for example, be bioavailable by oral administration whereas the parent drug is not.
  • the prodrug may also have better solubility than the active parent drug in pharmaceutical compositions.
  • prodrug a compound disclosed herein, which is administered as an ester (the "prodrug") to facilitate absorption through a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to a carboxylic acid (the active entity) once inside the cell where water-solubility is beneficial.
  • prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized in vivo to release the active parent compound.
  • the term “complement” refers to a oligonucleotide or polynucleotide that hybridizes by base-pairing, adenine to tyrosine and guanine to cytosine, to another oligonucleotide.
  • to "modulate" the activity of CBl means either to activate it, i.e., to increase its cellular function over the base level measured in the particular environment in which it is found, or deactivate it, i.e., decrease its cellular function to less than the measured base level in the environment in which it is found and/or render it unable to perform its cellular function at all, even in the presence of a natural binding partner.
  • a natural binding partner is an endogenous molecule that is an agonist for the receptor.
  • to "detect" changes in the activity of CBl or of a CBl sub-type refers to the process of analyzing the result of an experiment using whatever analytical techniques are best suited to the particular situation. In some cases simple visual observation may suffice, in other cases the use of a microscope, visual or UV light analyzer or specific protein assays may be required. The proper selection of analytical tools and techniques to detect changes in the activity of CBl or a CBl sub-type are well- known to those skilled in the art.
  • An "agonist” is defined as a compound that increases the basal activity of a receptor (i.e. signal transduction mediated by the receptor).
  • partial agonist refers to a compound that has an affinity for a receptor but, unlike an agonist, when bound to the receptor it elicits only a fractional degree of the pharmacological response normally associated with the receptor even if a large number of receptors are occupied by the compound.
  • An "inverse agonist” is defined as a compound, which reduces, or suppresses the basal activity of a receptor, such that the compound is not technically an antagonist but, rather, is an agonist with negative intrinsic activity.
  • antagonist refers to a compound that binds to a receptor to form a complex that does not give rise to any response, as if the receptor were unoccupied.
  • An antagonist attenuates the action of an agonist on a receptor.
  • An antagonist may bind reversibly or irreversibly, effectively eliminating the activity of the receptor permanently or at least until the antagonist is metabolized or dissociates or is otherwise removed by a physical or biological process.
  • a "subject” refers to an animal that is the object of treatment, observation or experiment.
  • Animal includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals.
  • “Mammal” includes, without limitation, mice; rats; rabbits; guinea pigs; dogs; cats; sheep; goats; cows; horses; primates, such as monkeys, chimpanzees, and apes, and, in particular, humans.
  • a "patient” refers to a subject that is being treated in order to attempt to cure, or at least ameliorate the effects of, a particular disease or disorder or to prevent the disease or disorder from occurring in the first place.
  • treating do not necessarily mean total cure or abolition of the disease or condition. Any alleviation of any undesired signs or symptoms of a disease or condition, to any extent can be considered treatment or therapy. Furthermore, treatment may include acts that may worsen the patient's overall feeling of well-being or appearance.
  • a “carrier” refers to a compound that facilitates the incorporation of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • DMSO dimethyl sulfoxide
  • a "diluent” refers to an ingredient in a pharmaceutical composition that lacks pharmacological activity but may be pharmaceutically necessary or desirable.
  • a diluent may be used to increase the bulk of a potent drug whose mass is too small for manufacture or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation.
  • a common form of diluent in the art is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mimics the composition of human blood.
  • an “excipient” refers to an inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition.
  • a “diluent” is a type of excipient.
  • Ri 3 and A are as defined above for Formula I.
  • R 4 , R 5 , and R 6 are appropriate radicals selected to obtain the desired A group.
  • compounds 2 5, 6, 10, 16, 26, 28 - 31, 40, 44, 46, 48, 51, and 58 can be prepared according to Scheme 1.
  • compound 2 can be prepared by using /j-butyl amine as H 2 N-Ri 3 and 2-pyridyl zinc bromide as R 6 ZnX.
  • Suitable heterocyclic 2-mercaptosubstituted carboxylic acids can be prepared as described in Blank et al, J. Med. Chem.
  • Ri 3 and A are as defined above for Formula I.
  • R 4 , R 5 , and R 6 are appropriate radicals selected to obtain the desired A group.
  • compounds 13-16, 18, 36-42, 54, 55, 57-60 can be prepared according to Scheme 2.
  • compound 13 can be prepared by using cyclohexyl amine a H 2 N-Ru and p-Cl-phenyl zinc iodide as R 6 ZnX.
  • heterocyclic 2-chloro carboxylic acids include commercially available 2-chloro nicotinic acid, 3-chloro-4-pyridine carboxylic acid, 4-chloronicotinic acid and 3-chloro-2-pyrazine carboxylic acid and those synthesized by methods described in Bredereck et al. Chem. Ber, 1962, 95, 956-963 and Krasovskiy et al., Angew. Chem. Int. Ed., 2006, 45, 2958- 2961, both of which are incorporated herein by reference in their entirety.
  • Scheme 3 illustrates one method of obtaining compound 7.
  • compounds 1, 3, 8, 9, 11, 19-25, 27, 32- 34, 43, 45, 49, 50, 52, 53, 61, 62, 82-84 can be obtained using a similar method.
  • Suitable iodo-or bromo benzoheterophenones can be obtained commercially, synthesized by the methods described in Reynolds et al, Tetrahedron, 2001, 57, 7765-7770, Liu et al., Org. Lett.
  • Ri a and A are as defined above for Formula I.
  • R 4 , R 5 , and R 6 are appropriate radicals selected to obtain the desired A group.
  • R 4 , R 5 , and R 6 compounds 4, 12, 17, 47, 56, 63, and 84 can be prepared according to Scheme 4 by using the appropriate heterocyclic 2-mercaptosubstituted carboxylic acids followed by the appropriate alkyl or aryl metal halide or amine and the desired amine in the last step.
  • Ri a and A are as defined above.
  • R 4 , R 5 , and R 6 are appropriate radicals selected to obtain the desired A group.
  • compounds 64, 68, 72, 78, 88, 92, and 96 can be prepared by using the appropriate heterocycle, amine, metal reagent/amine and amine. Further relevant synthetic methods can be found in Thompson et al, J. Org. Chem. 1988, 53, 2052-2055, which is incorporated herein by reference in its entirety.
  • Ru and A are as defined above for Formula I.
  • R 4 , R 5 , and R 6 are appropriate radicals selected to obtain the desired A group.
  • compounds 64-74, 85, 86, 88-91, 93- 95, 97 -101, and 104 can be prepared by using appropriate pyridines obtained either commercially or synthesized by magnesization of heterocycles via selective deprotonation as described in Liu et al, Org. Lett. 2006, 8, 617-619, incorporated herein by reference in its entirety, combined with the above mentioned 2-mercaptosubstituted carboxylic acids/esters followed by the appropriate reagents in the reaction sequence.
  • Scheme 7 illustrates one method of obtaining compound 102.
  • Other pyrazines such as compounds 87 and 102 can be obtained in a similar fashion following the methods described in PIe et al., J. Org. Chem. 1995, 60, 3781-3786, which is incorporated herein by reference in its entirety.
  • Pyrazines containing an amide side chain instead of sulfonamide side chain can be obtained by reacting the metalated species with a variety of alkyl isocyanates.
  • the dilithioketimine intermediate is obtained.
  • This intermediate can be in turn treated with suitable ⁇ -dihaloaryls or o-dihaloheteroaryls to obtain the thiazepine product.
  • ⁇ -deficient o-dihaloazaarenes constitute outstanding substrates for the reactions with ketimines, providing the thiazepine products in high yield simply by adding 1 equiv of a neat ⁇ -deficient ⁇ -dihaloazaarene to a reaction mixture containing a premade ketimine.
  • Ar 2 and D are as described above for Formula I.
  • A is an aryl or heteroaryl.
  • X is Br, Cl, F, OTf, or OTs.
  • Ari is a ⁇ -deficient azaaryl.
  • E is an electrophile including, but not limited to, isocyanates, acid chlorides, nitriles, tosyl cyanide or 1-cyanoimidazole, aldehydes or ketones, halogens or organic halides, carbon dioxide, Weinreb amides, tosyl azide, zinc chloride, tin chloride and trimethyl borate.
  • organometallic intermediates thus generated can be treated with convenient electrophiles to give substituted thiazepines.
  • substitution tactics afford a mixture of two or more regioisomeric products, the products can be easily separated from each other by one of the standard methods known in the art.
  • an electrophile can be added to a ⁇ - def ⁇ cient ⁇ -dihaloazaryl by a similar method as described above.
  • the resulting heteroaryl can be in turn be reacted with a ketimine to produce the desired thiazepine product.
  • Convenient substituents that can be introduced on the azaryl include, but are not limited to, nitrile or ester moieties. Introduction of such electron withdrawing groups on the rings of azaryls improves their reactivity towards ketimines. Also, at a later stage, these groups can be easily converted to other pertinent functional groups.
  • A is an alkyl, aryl, heteroaryl, or amino.
  • X is Br, Cl, F, OTf, or OTs.
  • Scheme 10 provides alkyl and amino substituted thiazepines by addition of 2 equiv of alkyllithiums, aryllithiums, heteroaryllithiums or 2 equiv of lithium amides to 2-cyanoarene thiols, giving rise to a ketimine, which can be reacted with ⁇ -dihaloaryl (or heteroaryl) to produce the desired thiazepine product.
  • the requisite 2-cyanoaryl thiols can be prepared for example by heating a 2-cyanobromoaryl (or heteroaryl) with mercaptoacetic ethyl ester in the presence of potassium t-butoxide and liquid ammonia (Brugelmans et al, Tetrahedron, 1983, 39, 4153, which is incorporated herein by reference in its entirety).
  • AI ⁇ , Ar 2 , B, and D are as described above for Formula I.
  • A is an alkyl, amino, aryl, or heteroaryl.
  • X is I, Br, or Cl.
  • Scheme 11 provides thiazepines by hydrolysis of ketimines followed by reacting the resulting ketoaryl thiol with a suitable o-aminohaloaryl (or heteroaryl). The cyclization to produce the thiazepines can be carried under copper catalysis (Bates et al, Org. Lett., 2002, 4, 2803, Kwong et al, Org.
  • ketones can also be prepared by other methods known in the art, for example by addition of organolithiums to mercaptoaryl(or heteroaryl)carboxylic acids (Bull. Chem.
  • the requisite iodoketones can be synthesized by one of the methods known in the art, for example by addition of organomagnesium reagents (Reynolds and Hermitage, Tetrahedron, 2001, 57, 7765, incorporated herein by reference in its entirety) to 2-iodo Weinreb amides (Brunette and Lipton, J. Org. Chem., 2000, 65, 5114, incorporated herein by reference in its entirety), by copper catalyzed aromatic Finkelstein reaction (Klapars and Buchwald, J. Am. Chem.
  • o-aminoaryl(or heteroaryl) thiol employed herein for the synthesis of thiazepines can be prepared by one of the methods known in the art, for example by reduction of ⁇ -nitroaryl(or heteroaryl) thiols (Foster and Reid, J. Am. Chem. Soc, 1924, 46, 1936, incorporated herein by reference in its entirety), by heating of 2- halonitroaryls(or heteroaryls) with sodium sulphide in water (Jain et al, Chem.
  • chloroketones can be reacted with o- aminoaryl(or heteroaryl) thiols to afford thiazepines. Presence of several strongly electron withdrawing groups (such as nitro or cyano groups) on the aromatic ring of the chloroketones (Jarret and Loudon, J. Chem. Soc, 1957, 3818, Gait and Loudon, J. Chem. Soc, 1959, 885, incorporated herein by reference in its entirety), or a ⁇ -deficient azaromatic chloroketone (Warmhof, Synthesis., 1972, 151, Shalaby, Phosphorus, Sulfur Relat. Elem., 2003, 775, 199, incorporated herein by reference in its entirety), is typically required.
  • strongly electron withdrawing groups such as nitro or cyano groups
  • terapéuticaally effective amount is used to indicate an amount of an active compound, or pharmaceutical agent, that elicits the biological or medicinal response indicated. This response may occur in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, and includes alleviation of the symptoms of the disease being treated.
  • One embodiment disclosed herein relates to a method of ameliorating or treating a disease or condition by administering to a subject a therapeutically effective amount of one or more compounds of Formula I.
  • the disease or condition can be selected from: obesity, metabolic syndrome, a metabolic disorder, hypertension, polycystic ovary disease, osteoarthritis, a deimatological disorder, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis, a sleep disorder, hyperlipidemic conditions, bulimia nervosa, a compulsive eating disorder, an appetite disorder, atherosclerosis, diabetes, high cholesterol, hyperlipidemia, cachexia, an inflammatory disease, rheumatoid arthritis, a neurological disorder, a psychiatric disorder, substance abuse (e.g., alcohol, amphetamines, amphetamine-like substances, caffeine, cannabis, ***e, hallucinogens, inhalents, nicotine, opioids, phencyclidine, phencyclidine
  • the therapeutically effective amount of a compound of Formula (I) is in a sufficient amount to ameliorate or treat said disease or condition by binding to a cannabinoid receptor (e.g., CB-I receptor).
  • the method can further include identifying a subject in need of ameliorating or treating said disease or condition.
  • T-cell mediated hypersensitivity disease comprising administering to a subject a therapeutically effective amount of a compound of Formula I.
  • methods include, but are not limited to methods such as a method of treating clinical manifestations in which cannabinoid receptor function is altered.
  • Some embodiments disclosed herein relate to a method for ameliorating or treating a disease or condition in which it would be beneficial to modulate the activity of a cannabinoid receptor, such as a CBl receptor, that can include administering to a subject a therapeutically effective amount of a compound of Formula I.
  • a cannabinoid receptor such as a CBl receptor
  • the neurological disorder can be schizophrenia, dementia, dystonia, muscle spasticity, tremor, psychosis, anxiety, depression, an attention deficit disorder, a memory disorder, a cognitive disorder, drug addiction, alcohol addiction, nicotine addiction, a neurodegenerative disease, multiple sclerosis, Alzheimer's disease, Parkinson's disease, epilepsy, Huntington's disease, Tourette's syndrome, cerebral ischaemia, cerebral apoplexy, craniocerebral trauma, stroke, spinal cord injury, pain, neuropathic pain disorder, viral encephalitis, and/or plaque sclerosis.
  • the disease or condition can be obesity, metabolic syndrome, appetite disorders, cachexia, high cholesterol, hyperlipidemia and/or diabetes.
  • the disease or condition can be of the gastrointestinal system such as emesis, nausea, gastric ulcers, diarrhea and/or intestinal disorders.
  • the disease or disorder can be an inflammation disease (e.g., rheumatoid arthritis, asthma, psoriasis).
  • an inflammation disease e.g., rheumatoid arthritis, asthma, psoriasis.
  • the disease or condition can be of the cardiovascular system such as hemorrhagic sock, septic shock, cirrhosis, atherosclerosis, and/or cardiovascular disorders.
  • the disease or condition can be of the reproductive system such as infertility and/or premature abortion.
  • the disease or condition can be of the visual system such as glaucoma, uveitis, retinopathy, dry eye and/or macular degeneration.
  • the disease or condition can be osteoporosis and/or ostepenia.
  • the disease or condition can be asthma and/or pleurisy.
  • the disease or condition can be cancer.
  • Another embodiment described herein relates to a method of ameliorating and/or treating drug and/or alcohol addiction comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
  • Still another embodiment described herein relates to a method for ameliorating or treating a disease or condition in which it would be beneficial to modulate the activity of a CBl receptor comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
  • Yet still another embodiment described herein relates to a method of ameliorating and/or treating obesity, comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
  • One still another embodiment described herein relates to a method of ameliorating and/or treating impaired cognition and/or a memory disorder comprising administering to a subject a therapeutically effective amount of a compound of Formula
  • Another embodiment described herein relates to a method of improving cognition or memory in a subject comprising administering to the subject a therapeutically effective amount of a compound of Formula (I)
  • Still another embodiment described herein relates to a method of ameliorating and/or treating inflammation due to an inflammatory disease comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
  • inflammatory diseases include rheumatoid arthritis, asthma, and psoriasis.
  • Any of the embodiments listed herein may further include identifying a subject in need of treatment or amelioration of any disease or condition identified herein.
  • FIG. 1 Another embodiments disclosed herein relate to a method of identifying a compound that treats or amerliorates any disease or condition identified herein in a subject, comprising identifying a subject suffering the disease or condition; providing the subject with at least one compound of Formula I, as defined herein; and determining if the at least one compound treats the disease or condition in the subject.
  • Some embodiments disclosed herein relate to a method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor in a subject that includes administering to the subject an effective amount of a compound of Formula I.
  • the cannabinoid receptor can be a CBl receptor.
  • inventions disclosed herein relate to a method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor comprising contacting a cannabinoid receptor with a compound of Formula I.
  • the cannabinoid receptor can be a CBl receptor.
  • Still other embodiments disclosed herein relate to a method of modulating or specifically inverse agonizing or antagonizing one or more cannabinoid receptors comprising identifying a subject in need of treatment or prevention and administering to the subject a therapeutically effective amount of a compound of Formula I.
  • Yet still other embodiments disclosed herein relate to a method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor that includes contacting a cannabinoid receptor with at least one test compound of Formula I; and determining any increase or decrease in activity level of the cannabinoid receptor so as to identify said test compound as an agonist, inverse agonist or antagonist of the cannabinoid receptor.
  • the cannabinoid receptor can be a CBl receptor.
  • the cannabinoid receptor can consists essentially of SEQ ID NO: 2.
  • the cannabinoid receptor can have at least 90% amino acid identity to SEQ ED NO: 2.
  • the cannabinoid receptor can have at least 85% amino acid identity to SEQ ID NO: 2.
  • the cannabinoid receptor can have at least 70% amino acid identity to SEQ ED NO: 2.
  • One embodiment disclosed herein relates to a method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor that includes culturing cells that express a cannabinoid receptor; incubating the cells or a component extracted from the cells with at least one test compound of Formula I; and determining any increase or decrease in activity of the cannabinoid receptor so as to identify said test compound as an agonist, inverse agonist, or antagonist of the cannabinoid receptor.
  • the cannabinoid receptor can be a CBl receptor.
  • the cannabinoid receptor can consists essentially of SEQ ID NO: 2.
  • the cannabinoid receptor can have at least 90% amino acid identity to SEQ ED NO: 2. In one embodiment, the cannabinoid receptor can have at least 85% amino acid identity to SEQ ED NO: 2. In another embodiment, the cannabinoid receptor can have at least 70% amino acid identity to SEQ ID NO: 2. [0173] Still another embodiment described herein relates to a method for identifying a compound which binds to a cannabinoid receptor comprising: labeling a compound described herein; with a detectable label; contacting the CB-I receptor with the labeled compound; and determining whether the labeled compound binds to the CB- 1 receptor. In an embodiment, the detectable label can be a radiolabel such as [ 3 H], [ 18 F], [ 11 C] and [ 125 IJ.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I as described above, and a physiologically acceptable carrier, diluent, or excipient, or a combination thereof.
  • composition refers to a mixture of a compound disclosed herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, intramuscular, intraocular, intranasal, intravenous, injection, aerosol, parenteral, and topical administration.
  • compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • Pharmaceutical compositions will generally be tailored to the specific intended route of administration.
  • physiologically acceptable defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
  • compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s).
  • suitable carriers or excipient(s) include butylene glycol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, s thereof.
  • Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, 18th edition, 1990, which is hereby incorporated by reference in its entirety.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, intraocular injections or as an aerosol inhalant.
  • compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.
  • compositions for use in accordance with the present disclosure thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., as disclosed in Remington's Pharmaceutical Sciences, cited above.
  • the agents disclosed herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds disclosed herein to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with pharmaceutical combination disclosed herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally, include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present disclosure are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactos
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • An exemplary pharmaceutical carrier for the hydrophobic compounds disclosed herein is a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • a common co-solvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • VPD co-solvent system which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of Polysorbate 80TM; the fraction size of polyethylene glycol may be varied; and other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone.
  • other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • salts may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free acids or base forms.
  • compositions suitable for use in the methods disclosed herein include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the exact formulation, route of administration and dosage for the pharmaceutical compositions disclosed herein can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al. 1975, in "The Pharmacological Basis of Therapeutics", Chapter 1, which is hereby incorporated by reference in its entirety).
  • the dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient's body weight, or 1 to 500 mg/kg, or 10 to 500 mg/kg, or 50 to 100 mg/kg of the patient's body weight.
  • the dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient. Where no human dosage is established, a suitable human dosage can be inferred from ED 50 or ID 5 o values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.
  • the daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg and 500 mg of each ingredient, preferably between 1 mg and 250 mg, e.g. 5 to 200 mg or an intravenous, subcutaneous, or intramuscular dose of each ingredient between 0.01 mg and 100 mg, preferably between 0.1 mg and 60 mg, e.g. 1 to 40 mg of each ingredient of the pharmaceutical compositions disclosed herein or a pharmaceutically acceptable salt thereof calculated as the free base, the composition being administered 1 to 4 times per day.
  • compositions disclosed herein may be administered by continuous intravenous infusion, preferably at a dose of each ingredient up to 400 mg per day.
  • the total daily dosage by oral administration of each ingredient will typically be in the range 1 to 2000 mg and the total daily dosage by parenteral administration will typically be in the range 0.1 to 400 mg.
  • the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety, which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compositions should be administered using a regimen, which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • compositions may, if desired, be presented in a pack or dispenser device, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • Compositions comprising a compound disclosed herein formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • Example 1 10-Phenyl-5-thia- 1 ,4, 11 -triazadibenzo[a,dlcycloheptene
  • Example 12 lO-Phenyl-5-thia-l Al l-triazadibenzo[ ⁇ . J]cvcloheptene-2.3-dicarboxylic acid bis-butylamide
  • n-butyl isocyanate 340 ⁇ L; 3 mmol.
  • the mixture was stirred at -78 0 C for 2 hours and then quenched by addition of a mixture of 4M HCl (200 ⁇ L), water (5 mL) and EtOH (5 mL) and the temperature raised to room temperature.
  • the mixture was neutralized by addition of NaHCO 3 (sat) and extraction performed with DCM.
  • R-SAT The functional receptor assay, Receptor Selection and Amplification Technology (R-SAT ® ), is used to investigate the pharmacological properties of CBl compounds described herein.
  • R-SAT is disclosed in U.S. Patent Nos. 5,707,798, 5,912,132, and 5,955,281, all of which are hereby incorporated herein by reference in their entirety, including any drawings.
  • NIH3T3 cells are grown in 96 well tissue culture plates to 70- 80% confluence. Cells are transfected for 16-20 h with plasmid DNAs using Polyfect (Qiagen Inc.) using the manufacturer's protocols. R-SATs are generally performed with 10 ng/well of receptor, 10 ng/well of Gqi5 (Conklin et al, Nature 1993 363:274-6) and 20 ng/well of ⁇ -galactosidase plasmid DNA. All receptor constructs are in the pSI-derived mammalian expression vector (Promega Inc).
  • the CBl receptor gene is amplified by PCR from genomic DNA using oligodeoxynucleotide primers based on the published sequence (GenBank Accession # X54937)
  • SEQ ED NO: 1 encodes a CBl receptor truncated after amino acid 417 (SEQ ID NO: 2).
  • the CB2 gene is cloned by performing a PCR reaction on mRNA from spleen.
  • the PCR product containing the entire coding sequence of the CB2 gene is cloned into an expression vector such that the CB2 gene is operably linked to an SV40 promoter.
  • the sequence of the CB2 gene (GenBank Accession #NM_001841) is provided as SEQ ID NO: 3 and the sequence of the encoded CB2 polypeptide is provided as SEQ ID NO: 4.
  • SEQ ID NO: 3 The sequence of the CB2 gene (GenBank Accession #NM_001841) is provided as SEQ ID NO: 3 and the sequence of the encoded CB2 polypeptide is provided as SEQ ID NO: 4.
  • Percent inhibition is calculated as the difference between the absorbance measurements in the absence of added ligand compared with that in the presence of saturating concentrations of ligand normalized to the absorbance difference for the reference ligand (SR141716), which was assigned a value of 100%.
  • the foregoing assay may be used to identify compounds which are agonists, inverse agonists or antagonists of a cannabinoid receptor.
  • the cannabinoid receptor used in the assay may be a CB 1 receptor.
  • the cannabinoid receptor used in the assay may consist essentially of SEQ ID NO: 2.
  • the cannabinoid receptor used in the assay may have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater than at least 99% amino acid identity with a full-length CBl receptor or a truncated CBl receptor of SEQ ID NO: 2.
  • the compounds disclosed herein are evaluated for their ability to bind to a CB 1 receptor.
  • the compounds are tested using a receptor binding assay and then determining of any change in GTPgamma S binding of transfected cells. The results indicate that the compounds are inverse agonists at the CBl receptor.
  • Membrane preparations Whole brains are harvested from Harlan Sprague Dawley rats and placed in 50 ml Falcon Tubes on ice. The volume is made up to 30 ml with ice-cold membrane buffer (20 mM HEPES, 6 mM MgCl 2 , 1 mM EDTA, pH 7.2). The Brains are homogenized with a Brinkmann Polytron PT3000 at 20,000 rpm for 40 s. The homogenate is spun at 1,000 x g for 10 min at 4 0 C to remove nuclei and cellular debris.
  • the supernatant is collected and re-centrifuged as previously before membranes are precipitated at 45,000 x g for 20 min at 4°C, resuspended in membrane buffer to a final concentration of 1 mg/ml, snap frozen as aliquots in liquid nitrogen and stored at - 80°C.
  • Membrane Binding - 10 ⁇ g of membranes are incubated in binding buffer (Ix DMEM with 0.1%BSA) in the presence of 3 nM radioligand ([ 3 H]SR141716A, Amersham Biosciences, Piscataway, NJ) and varying concentrations of ligands (total volume 100 ⁇ l in a 96 well plate).
  • binding buffer Ix DMEM with 0.1%BSA
  • 3 nM radioligand [ 3 H]SR141716A, Amersham Biosciences, Piscataway, NJ
  • Cells are filtered onto a 96 well GF/B filterplate (Packard Bioscience, Shelton, CT) and washed with 300 ml wash buffer (25mM HEPES, 1 mM CaCl 2 , 5 mM MgCl 2 , 0.25M NaCl) using a Filtermate 196 Harvester (Packard Instruments, Downers Grove, IL).
  • the filter plates are dried under a heat lamp before addition of 50 ⁇ l of scintillation fluid to each well (Microscint 20, Packard, Shelton, CT). Plates are counted on a Topcount NXT (Packard, Shelton, CT).
  • the CB 1 receptor binding assay may be used to identify compounds which are agonists, inverse agonists or antagonists of a cannabinoid receptor.
  • the cannabinoid receptor used in the assay may be a CBl receptor.
  • the cannabinoid receptor used in the assay may consist essentially of SEQ ID NO: 2.
  • the cannabinoid receptor used in the assay may have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater than at least 99% amino acid identity with a full-length CBl receptor or a truncated CBl receptor of SEQ ID NO: 2.
  • Rats Male, Sprague-Dawley rats (90-120 g) serve as subjects for these studies. Rats are fasted for a period of 16 hrs (water was always available). After the fasting period, test compounds are administered either intraperitoneally (ip) or orally (po). Immediately following compound administration, the rats are returned to their home cage. Following 30 min after compound administration, the rats are removed from their home cages and placed individually into clean cages with a pre-measured amount of food. Food weights are obtained (to the nearest 0.1 g) at various time points. Food consumption is monitored for a period of up to 2 hrs (i.e., 2.5 hr after test compound administration).
  • mice Male, NSA mice (15-20 g) serve as subjects for these studies. Baseline nociceptive thresholds are assessed using the warm water tail flick test. Briefly, the distal 1/3 to 1 A of the tail is immersed in a 52 0 C water bath and the time (to the nearest 0.1 sec) until the mouse removed its tail (i.e., "flicks") from the water is recorded (i.e., tail flick latency). Mice are then injected ip with either vehicle or with various doses of the CBl agonist CP 55,940 and tail flick latencies are recorded for a period of up to 3 hr. A maximum latency of 10 sec is employed in order to prevent tissue damage.
  • mice are pretreated with either vehicle or with a test compound 30 min prior to CP55,940.
  • CP55,940 (1 mg/kg) is administered subcutaneously, and the test compound is administered intraperitoneally. Tail flick latencies are then obtained at various time points for a period of up to 2 hr.
  • the vehicle for both compounds is 1:1: 18 cremphor:ethanol:saline.
  • mice Male, NSA mice (15-20 g) serve as subjects for these studies.
  • mice are pretreated with either vehicle or with test compound 30 min prior to CP55,940. Core body temperatures are then obtained at various time points following CP 55,940 administration. Core body temperature (to the nearest 0.1 0 C) is obtained by rectal probe.
  • Rats Male, obese Zucker rats (400-500 g) serve as subjects for these studies. Rats are housed individually and have access to food and water ad libitum. Rats are allowed to acclimate to the vivarium for a period of 3 days, during which body weight and consumption of food and water is monitored. Rats are weighed daily at 1500 hr and then injected with either vehicle or with various doses of the test compound. Daily food and water intakes are also monitored. Food and water bottles are weighed at the time body weights are recorded (i.e., 1350 hr). Vehicle or compound is administered daily for a period of up to 15 days. The test compound attenuates the food and water intake of the rats. Moreover, the attenuation of the food and water intake is dose-dependent.
  • Subjects are male, C57 BK/6 mice, weighing 15-20g upon arrival. Animals are housed 8 per cage with food and water available ad libidum. Animals are housed on a 12 hr light cycle (lights on 6 am) for 4-7 days prior to behavioral testing.
  • Equipment Novel object recognition (NOR) is conducted in a novel environment consisting of a white plastic tub measuring 45.7 x 33.7 x 19 cm. Prior to each trial the bottom of the tub is covered with a piece of plastic lined bench top paper. There are two sets of identical objects chosen so that when given a opportunity to explore, mice would evenly divide exploration time between the objects. "A” objects are yellow, ceramic, 12-sided ramekins measuring 4 cm high x 7 cm diameter. "B” objects are 8 X 8 x 4 cm stainless steel, 4-sided ramekins.
  • Procedure At the beginning of each test day, animals are placed in groups of 6 into clean cages. Testing is conducted in three phases: acclimation, sample and test. For acclimation, each group of six mice is placed collectively into the NOR chamber and allowed to explore freely for 30 min. After acclimation animals are injected (dose and pretreatment time varied by test drug) and placed back into the cages to wait the pre-treatment interval. After the pre-treatment time elapsed, each mouse is placed, one at a time into the NOR chamber, into which two identical objects have been placed ("A" or "B" objects described above). Objects are placed on diagonal corners of the long axis of the arena approximately 5 cm from the walls, while subjects are placed into one of the neutral corners (alternating across subjects).
  • Each mouse is allowed to explore the chamber and the objects for 3 min., and the time spent exploring at each position is recorded. Directly sniffing or touching the object is recorded as exploration. After 3 min., each mouse is removed from the arena and placed back into its cage.
  • the test phase was conducted 1 or 2 hours after the sample phase. During test, one familiar object (seen during sample) and one novel object are placed into the chamber in the same positions used during the sample phase, and each mouse is allowed 3 min to explore.
  • the test sessions are recorded on video and scored by an observer blind to each subject's treatment condition. Any time spent directly sniffing or touching an object was counted as exploration.
  • the object serving as the novel object and the position where the novel object is placed are counterbalanced across subjects. Prior to each trial (acclimation, sample and test), all equipment is wiped with a Clorox wipe and bench paper (cut to fit) is placed in the bottom of the chamber. The procedure is shown below in Scheme 13.
  • mice/ Group Acclimation - 30 min. (6 mice/ Group)
  • Subjects are male, Sprague-Dawley rats purchased from Harlan Laboratories, weighing 250-275 g upon arrival. Prior to surgery animals are housed two per cage. All subjects have free access to food and water available for the duration of the study. Animals are housed on a 12 hr light cycle (lights on 6 am), and are acclimated to vivarium conditions for a minimum of one week prior to surgery. All experiments are conducted in accordance with NIH Guidelines for the Care and Use of Laboratory Animals and are approved by the Institutional Animal Care and Use Committee at ACADIA Pharmaceuticals, Inc.
  • Animals are placed in the stereotaxic instrument with the incisor bar at -3.2 mm and a hole is drilled in the skull over the substantia nigra according to the atlas of Paxinos and Watson (1997): A/P -5.2 mm, M/L - 2.1 mm.
  • a computer-controlled microsyringe is lowered to -8.2 mm from bregma.
  • 8 ⁇ g of 6-hydroxy-dopamine in 4 ⁇ l of saline with 0.2% ascorbic acid is infused over 5 min, and 1 min is allowed for diffusion before the syringe is removed and the incision closed. Animals are given a minimum of 15 days after surgery before any behavioral assessment.
  • Rotational Behavior All animals are assessed for rotational behavior in rotometers purchased from San Diego Instruments, Inc. For each behavioral session, subjects are placed in the rotometers and allowed thirty minutes for acclimation. After 30 min., subjects are injected with either the dopamine agonist apomorphine (0.05, 0.16 or 0.5 mg/kg ip in saline with 0.2% ascorbic acid) or the cannabinoid 1 receptor inverse agonist Compound II, N-(butyl)-l l-(4-chlorophenyl)-dibenzo[b,f,][l,4]thiazepine-8- carboxamide, (3 mg/kg in sesame oil).
  • dopamine agonist apomorphine 0.05, 0.16 or 0.5 mg/kg ip in saline with 0.2% ascorbic acid
  • cannabinoid 1 receptor inverse agonist Compound II N-(butyl)-l l-(4-chlorophenyl)-dibenzo
  • Compound II When subjects receive combinations of the two treatments, Compound II is injected 30 minutes prior to apomorphine. After treatment, rotations is measured for 60 min. Subjects are then removed from the rotometers and returned to their home cages. All animals receive all three doses of apomorphine, and the combination of Compound II with both 0.05 mg/kg and 0.16 mg/kg apomorphine. A minimum of 2 days separated test days.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Addiction (AREA)
  • Psychiatry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyrane Compounds (AREA)

Abstract

Disclosed herein is a compound of Formula (I). Also disclosed herein is a method of modulating the activity of a cannabinoid receptor using a compound of Formula (I). Furthermore, disclosed herein is a method of treating a disease or condition that would be alleviated, improved or prevented by administration of a compound that modulates a cannabinoid receptor comprising identifying a subject in need thereof and administering to said subject a therapeutically effective amount of a compound of Formula (I). Also disclosed herein are pharmaceutical compositions comprising a compound of Formula (I).

Description

CBl-MODULATING COMPOUNDS AND THEIR USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. Provisional Application No. 60/852,579, entitled "CBl-MODULATING COMPOUNDS AND THEIR USE", filed October 17, 2006; which is incorporated by reference in its entirety including all drawings.
BACKGROUND OF THE INVENTION Field of the Invention
[0002] This invention relates to the fields of organic chemistry, pharmaceutical chemistry, biochemistry, molecular biology and medicine. In particular it relates to compounds that modulate the activity of the human cannabinoid receptor (CBl), and to the use of the compounds for the treatment and prevention of diseases and disorders related to CB 1.
Description of the Related Art
[0003] The cannabinoids, which are bioactive lipids, naturally found in the cannabis sativa (marijuana) plant, have been used recreationally and therapeutically for at least 5000 years. In addition to their well-documented effects on mood, cannabinoids (often in the form of marijuana) have been prescribed to treat nausea, pain, migraine, epilepsy, glaucoma, hypertension, cachexia and pain associated with childbirth. Two cannabinoid receptors, CBl and CB2, have been identified. Both are members of the G protein-coupled receptor superfamily, and are negatively coupled through Gi protein. The CB2 receptor has 44% sequence similarity to the CBl receptor.
[0004] The CB 1 receptor, unlike the CB2 receptor, is highly expressed in the central nervous system, mostly presynaptically. Indeed, the CBl receptor is present in the brain at higher levels than many other GPCRs. It is found in the cortex, cerebellum, hippocampus, and basal ganglia (reviewed in Brievogel and Childres, 1998). In addition, the CBl receptor has also been detected in sperm, the prostate gland, and other peripheral tissues (including structures of the eye). The CB2 receptor is present in the cells of the immune system (spleen, thymus), testis, and lung.
[0005] The CBl receptor is believed to be responsible for the appetite stimulating properties and habituation associated with cannabinoid use. The CBl receptor antagonist, SR141716 (rimonabant, Acomplia; Sanofϊ-Aventis) has shown efficacy in late-stage clinical trials for obesity and nicotine dependence, with no psychotropic effects. The compound has been shown to reduce both food intake and adipose tissue (by a mechanism independent of food intake). Use of SR141716 in animal models suggests additional use of CB 1 receptor antagonists and inverse agonists for the treatment of alcohol addiction, opiate addiction, ***e addiction, anxiety, and septic shock. Interestingly, mice null for the CBl gene also display impaired ***e self- administration, and less severe withdrawal from morphine addiction compared to wild- type mice. In addition, CBl knockout mice also display increased bone mineral density, and both CBl knockout mice and mice treated with CB antagonists are resistant to bone loss in a model for osteoporosis. Other animal models indicate a use for CBl receptor antagonists and inverse agonists for the prevention of premature spontaneous abortion.
[0006] Cannabinoid signaling is hyperactive in animal models of several diseases suggesting that cannabinoids either have a protective role (e.g., CBl agonists may be therapeutic) or are involved in the pathology of these diseases (e.g., CBl antagonists or inverse agonists may be therapeutic). These include Parkinson's disease, Alzheimer's disease, multiple sclerosis, epilepsy, and intestinal disorders. In addition, the levels of endogenous cannabinoids and CBl receptors are elevated in the liver and blood of patients with cirrhosis of the liver. Moreover, cannabinoid levels have been shown to be elevated in the cerebrospinal fluid of a patient with stroke, as well as in the brains of depressed suicide victims. Endogenous cannabinoids have also been shown to be higher in the cerebrospinal fluid of drug-naive paranoid schizophrenics compared to normal patients; interestingly, schizophrenic patients treated with atypical but not typical antipsychotics also exhibit higher CSF levels of anandamide. Additionally, the CBl gene is located in a chromosomal region that has been linked to schizophrenia. Moreover, high levels of the endogenous cannabinoid, anandamide, are correlated with premature abortion and failure of in vitro fertilization. Finally, activation of CB receptors by an anandamide analogue has been shown to reduce sperm fertilizing capacity by 50%.
[0007] Selective activation of CBl receptors by agonists or partial agonists may also be used to treat a number of disorders. Some patients in clinical trials of the CBl antagonist, SR141716A, have reported diarrhea and nausea, suggesting that an agonist would alleviate those symptoms. THC (tetrahydrocannabinol; active cannabinoid in Cannabis sativa) has been shown to improve mobility and alleviate pain in patients with multiple sclerosis. Other promising results for cannabinoids have been shown in clinical trials for Tourette's syndrome, Parkinson's disease, glaucoma, and pain. Finally cannabinoids have been shown to inhibit cancer growth, angiogenesis, and metastasis in animal models.
SUMMARY OF THE INVENTION
[0008] Disclosed herein is a compound of Formula (I) in which A, B, D X, Y, Aη and Ar2 are defined herein.
Figure imgf000004_0001
(I) [0009] Also disclosed herein are pharmaceutical compositions comprising a compound of Formula (I). Furthermore, disclosed herein is a method of treating a disease and/or condition that would be alleviated, ameliorated, and/or treated by administration of a compound that modulates a cannabinoid receptor comprising administering to a subject a therapeutically effective amount of a compound of Formula (I). Also disclosed herein is a method of modulating the activity of a cannabinoid receptor using a compound of Formula (I). DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS [0010] One embodiment described herein relates to a compound of Formula (I):
Figure imgf000005_0001
as a single isomer, a mixture of isomers, a racemic mixture of isomers, or a pharmaceutically acceptable salt, solvate, metabolite, prodrug, or polymorph thereof.
[0011] X can be selected from O, S, S=O, SO2, NRi, NC≡N, NC(=Z)R,, NC(=Z)NRiaRib, CRiaRib, C-O, C=CRiaRit>, and SiRuRib- Y can be -N(R2) — or - C(RiR2) = where the symbol ^^ represents a single or double bond, where when =^= is a double bond, R2 is absent. A can be selected from halogen, -NRiaRib, and -N=CRiaRib; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, (cycloalkyl)alkyl, (cycloalkenyl)alkyl, (cycloalkynyl)alkyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono- substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, sulfonyl, and -(CH2)0-4-C(=Z)-ORi; provided that A cannot be a substituted or unsubstituted piperazine.
[0012] Ari and Ar2 can be separately selected from aryl and heteroaryl, each optionally substituted with one or more radical selected from halogen, hydroxyl, nitro, - CN, -Q=Z)R1, -C(=Z)OR,, -C(=Z)NR,aRlb, -C(=Z)N(R,)NRlaRlb, -C(=Z)N(R,)N(R1)C(=Z)R1, -C(R,)=NR,, -NR13Rn,, -N=CR,aRlb) -N(R1)-C(=Z)R1, -N(R,)-C(=Z)NRlaRlb, -S(O)NRlaRlb, -S(O)2NRlaRlb, -N(R,)-S(=O)R,,
-N(Ri)-S(=O)2Ri, -ORi, -SRi, and -OC(=Z)Ri; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, haloalkyl, and haloalkoxy; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, and sulfonyl; provided that at least one of Aη and Ar2 is an optionally substituted heteroaryl.
[0013] D can be part of Ar1 and is selected from CRi, NR2, S, and O. B can be attached to Ari on the carbon adjacent to D and separated from Y by three atoms, wherein one of the three atoms is D, and is selected from halogen, hydroxyl, nitro, -CN, -C(=Z)R|, -CC=Z)OR1, -C(=Z)NRlaRlb, -C(=Z)N(R,)NRlaRlb, -C(=Z)N(R,)N(R,)C(=Z)R,, - C(RO=NR1, -NR,,Rlb, -N=CRlaR.b, -N(R,)-C(=Z)R,, -N(R,)-C(=Z)NRlaRlb, - S(O)NRlaRlb, -S(O)2NRlaRlb, -N(R,)-S(=O)R,, -N(R,)-S(=O)2Ri, -OR1, -SR1, and -OC(=Z)Ri; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, haloalkyl, and haloalkoxy; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, and sulfonyl.
[0014] R1; Rla and Rib can be each' independently selected from hydrogen, halogen, -C(=Z)R3, -C(=Z)OR3, and -C(=Z)NR3aR3b; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, and haloalkyl; and mono- substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, -(CH2)0-7-OR3, - (CH2)o-7-SR3, and -(CH2)o-7-NR3aR3b; or Ria and Rlb can be taken together to form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms or an unsubstituted or substituted carbocyclyl having 3 to 9 carbon atoms. R2 can be absent or is selected from hydrogen; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, and alkynyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl. R3, R3a, and R3b can be each independently selected from: hydrogen; mono-substituted, poly- substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, aryl.lieteroaryl, and heteroalicyclyl. Z can be O or S.
[0015] In some embodiments, B is not selected from -CF3, phenyl, -OS(O)2-
CF3, methyl, -CN, halogen, and
Figure imgf000007_0001
when A is a substituted or unsubstituted heteroalicyclyl containing at least one nitrogen, cycloalkyl, cycloalkenyl, phenyl, heteroaryl, or -NRuRib- In an embodiment, B is not halogen when A is substituted or unsubstituted aryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroaryl, halogen, or substituted or unsubstituted sulfenyl; X is -NH; and Y is -N=. In some embodiments, when X is O or -NCH3 and Y is -N=, then B cannot be -C(=Z)OH, - C(=Z)Me or -C(=Z)Et. In an embodiment, when X is CR^Rib and A is phenyl, then B cannot be NH2. In some embodiments, when both Ari and Ar2 are pyridinyl rings, then X cannot be NRj in which Ri is selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, trihalomethyl and hydroxyalkyl. In an embodiments, when X is S or NRi, wherein Ri is hydrogen or alkyl, then A cannot be a phenyl ring substituted at the para-position with -CO2H or CO2(alkyl). In some embodiments, when X is CRiaRib or C=CRiaRib, then R)3 and Rib cannot be a cycloalkyl, cycloalkenyl or piperazine ring, and Ri a and R^ cannot be taken together to a form a cycloalkyl, cycloalkenyl or piperazine ring. In an embodiment, when X is NRi, wherein Ri is hydrogen, -C(=O)H or -C(=O)CF3, then A cannot be a phenyl ring substituted at the para-position with a nitro group. In some embodiments, when X is NRi and A is aryl, then B cannot be alkyl, alkoxy, hydroxy, or an acid salt thereof. In an embodiment, when X is NRi and Y is -N(R2)^"=, wherein =^= represents a single bond, then B cannot be alkyl or alkoxy.
[0016] In some embodiments, Ri3 and Rib can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms and substituted with subtituents selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino. In other embodiments, Ru and Rib can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from:
Figure imgf000008_0001
wherein R4 and R5 are each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxyl, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino. In still other embodiments, Ri3 and R^ can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from:
Figure imgf000008_0002
[0017] In some embodiments, Rib can be hydrogen. In other embodiments,
Rib can be Ci-3alkyl.
[0018] In an embodiment, X can be S (sulfur). In an embodiment, Y is - N(R2) ^=, wherein ^^ represents a double bond and R2 is absent.
[0019] In some embodiments, B can be selected from halogen, -C(=Z)Ri, -C(=Z)ORi, -C(=Z)NRi.Rib, -C(=Z)N(R,)NR,aRlb, -C(=Z)N(R,)N(R,)C(=Z)R,, - C(RO=NR1, -NRi,R,b, -N=CRiaR,b, -N(R,)-C(=Z)R,, -N(R1)-C(=Z)NR,aRlb, - S(O)NR,aRib, -S(O)2NRlaRlb, -N(R,)-S(=O)R,, -N(Ri)-S(=O)2R,, and -OC(=Z)R,; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: heteroaralkyl and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: aryl, heteroaryl, aralkyl, and heteroalicyclyl. In other embodiments, B can be selected from hydroxyl, -CC=Z)OR1, -C(=Z)N(R,)NR,,R,b, -CC=Z)N(R1)N(R1)CC=Z)R1, -C(R,)=NR,, -NRlaRlb, - N=CR,aRib, -S(O)NRi,Rib, -N(Ri)-S(O)Ri, -N(Ri)-S(O)2Ri, and -OC(=Z)R,; mono- substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, and haloalkoxy; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfϊnyl, and sulfonyl. In one embodiment, B can be selected from aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, halogen, -C(=Z)R,, -C(=Z)ORi, -C(=Z)NRiaR,b, -C(=Z)N(R,)NRiaR,b, -C(=Z)N(R,)N(R,)C(=Z)R,, -C(Ri)=NR1, -NRlaRlb, -N=CRlaRlb) -N(R,)-C(=Z)Ri, -N(R,)-C(=Z)NR,,R,b, -S(O)NRlaRlb, -S(O)2NRlaRlb, -N(R,)-S(O)R,, -N(Ri)-S(O)2Ri, and -OC(=Z)Ri, wherein any member of said group can be substituted or unsubstituted. In another embodiment, B can be selected from cycloalkyl, cycloalkenyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, hydroxyl, sulfenyl, sulfϊnyl, sulfonyl, haloalkoxy, -C(=Z)ORi, -C(=Z)N(R1)NRiaRib, -C(=Z)N(R,)N(R,)C(=Z)Ri, -C(RO=NR1, -NR,,R,b, -N=CRlaRIb, -S(O)NR,,R,b, -N(RO-S(O)R1, -N(RO-S(O)2R1, and -OC(=Z)Ri, wherein any member of said group can be substituted or unsubstituted. In still another embodiment, B can be selected from cycloalkyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted. In yet still other embodiments, B can be an unsubstituted or substituted heteroaryl selected from:
and
Figure imgf000010_0001
. In one embodiment, B can be an optionally substituted phenyl. In an embodiment, the optionally substituted phenyl can be substituted with a CM alkyl.
[0020] In some embodiments, B can be -C(=Z)NRiaRib In one embodiment, when B is -C(=Z)NRiaRib> Ria can be selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH2)0-7-NR3aR3b, wherein any member of said group can be substituted or unsubstituted. In an embodiment, Ria can be selected from hydrogen, alkyl, alkoxy, aryl, aralkyl, heteroaryl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted. In some embodiments, Ri a can be an optionally substituted heteroaryl or heteroaralkyl. hi an embodiment, when B is -C(=Z)NRiaRib and Ri3 is hydrogen, alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH2)o-7-NR3aR3b, Rib can be hydrogen or methyl, hi an embodiment, the optionally substituted heteroaryl or
heteroaralkyl can be selected from:
Figure imgf000010_0002
Figure imgf000010_0003
, wherein Q is oxygen or sulfur, and in some embodiments, n can be 1 or 2. hi more particular embodiments, the optionally substituted heteroaralkyl can be
Figure imgf000010_0004
; and in some embodiments, n can be 1 or 2. [0021] In other embodiments, B can be -C(=Z)R, or -C(=Z)ORi. In one embodiment, B can be -C(=Z)Ri and Ri can be selected from alkyl, cycloalkyl, aralkyl, halogen. In an embodiment, B can be -C(=Z)Ri and Rj can be an alkyl. In an embodiment, B can be -C(=Z)ORi and Rj can be alkyl or aralkyl.
[0022] In still other embodiments, B can -C(=Z)N(Ri)N(Ri)C(=Z)Ri or -N(R,)-C(=Z)NRi,Rib. In an embodiment, -C(=Z)N(Ri)N(R,)C(=Z)R, can be
Figure imgf000011_0001
, wherein n is 0 or 1. In certain other embodiments, B can be -N(Ri)-C(=Z)NRiaRib and Ri is hydrogen and Ria is alkyl or aralkyl. In any of the embodiments discussed in the present paragraph, Rn, can be hydrogen.
[0023] In yet still other embodiments, B can be selected from
Figure imgf000011_0002
-N(Ri)-C(=Z)Ri, and -OC(=Z)Ri. In certain embodiments, B can be -C(Ri)=NRi, -N(Ri)-C(=Z)Ri, and -OC(=Z)Ri wherein at least on Ri is hydrogen or alkyl and at least one Ri is selected from alkyl, aryl, and aralkyl.
[0024] In some embodiments, B can be -N(Ri)-S(=O)Ri or -N(Ri)-S(=O)2Ri. In an embodiment, B can be -N(Ri)-S(=O)Ri or -N(Ri)-S(=O)2Ri and Ri can be hydrogen, aralkyl, or heteroaryl.
[0025] In other embodiments, B can be -S(O)NRiaRib or -S(O)2NRiaRib. In an embodiment, B can be -S(O)NRiaRib or -S(O)2NRi3RIb and Ria can be selected from alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl. In some embodiments, B can be - S(O)2NRiaRib and Ria can be selected from alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl. In an embodiment, B can be -S(O)2NRiaRib and Ria can be alkyl or heteroaryl. In any of the embodiments discussed in the present paragraph, R^ can be hydrogen.
[0026] In one embodiments, B can be -S(O)NRiaRib, -S(O)2NR,aRib, -C(=Z)NRiaRib or -C(=Z)N(Ri)NRiaRib and Rt; R)a and Rib can each independently selected from:
Figure imgf000012_0001
wherein: n can be an integer selected from O, 1, 2, 3, 4, 5, 6 or 7 defining the number of optionally substituted carbon atoms;
Q can be selected from -N(R4)-, O and S;
R4 and R5 each each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino; and
R6, Rδa, Rόb, Rόc, and R61I can each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S- sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino; or wherein the substituents selected from R6, R62, Rόb, R60, and Rod can be taken together to form a cycloalkyl, cycloalkenyl, cycloalkynyl, or heteroalicyclyl ring with one or more adjacent members of said group consisting of R6, R63, Rόb, R6c, and Rod- In an embodiment discussed in this paragarph, B can be -C(=Z)NRiaRib In an embodiment discussed in this paragarph, B can be -C(=Z)NRiaRib and n can be 0, 1, or 2. In any of the embodiments discussed in the present paragraph, R^ can be hydrogen. In an embodiment discussed in this paragarph, B can be -C(=Z)NRiaRib and Rib can be hydrogen. In an embodiment discussed in this paragarph, B can be -C(=Z)NRiaRib, Rib can be hydrogen, and n can be 0, 1, or 2.
[0027] In some embodiments, Ri, R|a, R2a, R2, R3, R3a, and R3b can be each independently selected from aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, or (heteroalicyclyl)alkyl and are substituted with zero to five substituents, wherein each substituent is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, . carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino. In an embodiment, Ri; Rla and R^ can be independently selected from f hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl. In embodiment, Ri and Ri3 can be independently selected from of alkyl, cycloalkyl, heteroaryl, heteroalicyclyl and heteroaralkyl; and R^ can be hydrogen.
[0028] In one embodiment, A can be an aryl, heteroaryl, or heteroalicyclyl, and is substituted with zero to five substituents, wherein each substituent is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino, and protected amino. In an embodiment, A can be selected from halogen, -NRi3Rn,, and -N=CRI3RH,; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, (cycloalkyl)alkyl, (cycloalkenyl)alkyl, (cycloalkynyl)alkyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, sulfonyl, and -(CH2)0-4-C(=Z)-ORi . In an embodiment, A can be an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent can be independently selected from alkyl, alkoxy, ester, cyano, and halogen. In some embodiments, the heteroaryl can be substituted or unsubstituted thiophene or substituted or unsubstituted pyridine. In other embodiments, the aryl can be an unsubstituted or substituted phenyl (e.g., 2-, 3-, 4-, 2-,3-, 2-,4- substituted phenyl). In some embodiments, A can be an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent is independently selected from alkyl, alkoxy, ester, cyano, and halogen. In an embodiment when A is substituted phenyl, the phenyl can be substituted with a halogen, methoxy, or cyano group. In some embodiments, A can be alkyl or aryl. In other embodiments, A can be cycloalkyl.
[0029] In some embodiments, X can be selected from S, S=O, and SO2; Y can be -N(R2) =÷ or -C(RiR2) ^^; the symbol ^-= represents a single or double bond, where when ^= is a double bond, R2 is absent; A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstiruted; B, C, D, E, F, G and I can be separately selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, halogen, hydroxyl, nitro, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, -CN, -C(=Z)Ri, -C(=Z)ORU -C(=Z)NR,aRib, -C(=Z)N(R,)NRlaRlb, -C(=Z)N(R,)N(R,)C(=Z)R,, -C(Ri)=NRi, -NRlaRib, -N=CR,aR,b, -N(R,)-C(=Z)Ri, -N(R,)-C(=Z)NRiaRlb, -S(O)NRlaRlb, -S(O)2NRlaR,b, -N(R,)-S(=O)R,, -N(Ri)-S(=O)2Ri, -ORi, -SRi, and -OC(=Z)R)t wherein any member of said group can be substituted or unsubstiruted except for hydrogen; B can be selected from -C(=Z)NRiaRib, -C(=Z)N(R,)NRlaRib, -C(=Z)N(R,)N(Ri)C(=Z)Ri, and -C(Ri)=NR1, wherein any member of said group can be substituted or unsubstiruted; Z can be O or S; Ri, Ri3 and Rib can each independently selected from: hydrogen, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, -(CH2)0.7-OR3, -(CH2)0-7-SR3, -(CH2)0-7-NR3aR3b, haloalkyl, -C(=Z)R3, -C(=Z)OR3, and -C(=Z)NR3aR3b, wherein any member of said group can be substituted or unsubstiruted except for hydrogen; or Ria and Rib can be taken together to form an unsubstiruted or substituted heteroalicyclyl having 2 to 9 carbon atoms or an unsubstiruted or substituted carbocyclyl having 3 to 9 carbon atoms; R2 can be absent or is selected from: hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted except for hydrogen; and R3i R3a, and R3b can each independently selected from: hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, and (heteroalicyclyl)alkyl, wherein any member of said group can be substituted or unsubstituted except for hydrogen. In one embodiment, Z can be O (oxygen). In another embodiments, A can be selected from C3- Ci2alkyl (e.g., n-propyl), C4-Ci2alkyl (e.g., n-butyl), cycloalkyl (e.g., cyclohexyl), aryl (e.g., substituted or unsubstituted phenyl), and heteroaryl (e.g., thiophene and pyridine), wherein any member of said group can be substituted or unsubstituted. In yet another embodiment, Z can be O (oxygen) and A can be selected from C3-Ci2alkyl (e.g., n- propyl), C4-Ci2alkyl (e.g., n-butyl), cycloalkyl (e.g., cyclohexyl), aryl (e.g., substituted or unsubstituted phenyl), and heteroaryl (e.g., thiophene and pyridine), wherein any member of said group can be substituted or unsubstituted.
[0030] In some embodiments, A can be selected from C3-Ci2alkyl (e.g., n- propyl), C4-Ci2alkyl (e.g., n-butyl), cycloalkyl(e.g., cyclohexyl), aryl(e.g., substituted or unsubstituted phenyl), heteroaryl(e.g., thiophene and pyridine), heteroalicyclyl (e.g., piperidine), halogen, -NRuRib, and -(CH2)0-4-C(=Z)-ORi . In other embodiments, A can be selected from C3-Ci2alkyl (e.g., n-propyl), C4-Ci2alkyl (e.g., n-butyl), cycloalkyl(e.g., cyclohexyl), aryl(e.g., substituted or unsubstituted phenyl), heteroaryl(e.g., thiophene and pyridine), heteroalicyclyl (e.g., piperidine), halogen, -NRiaRib, and -(CH2)0.4-C(=Z)-ORi; and X can be S (sulfur). In an embodiment, A can be -NR^Rib wherein Ri3 is an aryl (e.g., optionally substituted phenyl) and R^ is hydrogen. In certain other embodiments, A can be -NRuRib wherein Ria is a phenyl group substituted with a halogen and R^ is hydrogen. In an embodiment, A can be C3-Ci2alkyl (e.g., n-propyl), C4-Ci2alkyl (e.g., n- butyl). In certain other embodiments, A can be cycloalkyl (e.g., cyclohexyl). In other certain embodiments, A can be aryl (e.g., substituted or unsubstituted phenyl). In an embodiment, the aryl can be an unsubstituted or substituted phenyl (e.g., 2-, 3-, 4-, 2-,3-, 2-,4- substituted phenyl) In certain other embodiments, A can be heteroaryl (e.g., optionally thiophene or optionally substituted pyridine). In some embodiments, A is not C3-, C4-, C5-, C6-, C7-, C8-, C9-, Ci0-, Cn-, Ci2 alkyl. In other embodiments, A is not C4-, C5-, C6-, C7-, C8-, C9-, C10-, Cn-, C12 alkyl. In still other embodiments, A is not cycloalkyl. In some embodiments, A is not aryl. In other embodiments, A is not heteroaryl. In still other embodiments, A is not heteroalicyclyl. In yet still embodiments, A is not halogen, -NRuRib. In some embodiments, A is not -(CH2)0-4-C(=Z)-ORi. [0031] In some embodiments, A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)O-4- C(=Z)-ORi; X can be S (sulfur); and Y can be -N(R2) =-^ wherein the symbol ^^ represents a double bond and R2 does not exist. In some embodiments, A can be selected from C3-Ci2alkyl, C4-Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, - NRlaRib, and -(CH2)0-4-C(=Z)-ORi; X can be S; Y can be -N(R2) — wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib- In an embodiment, A can be selected from C3-Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)o-4-C(=Z)-ORi ; X can be S; Y can be -N(R2) ^^ wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib. In certain other embodiments, A can be an aryl or a heteroaryl group; X can be S; Y can be - N(R2) ^^ wherein the symbol =^÷ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib- In an embodiment, A can be a cycloalkyl, a heteroalicyclyl, or - NRiaRib group; X can be S; Y can be -N(R2)=-^ wherein the symbol ^=-= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib- In some embodiments X can be S; Y can be -N(R2) =^ wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein R]3 can be selected from the group consisting of alkyl, alkoxy, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH2)o-7-NR3aR3b, wherein any member of said group can be substituted or unsubstituted.
[0032] In some embodiments, A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)O-4- C(=Z)-ORi; X can be S; Y can be -N(R2)=-^ wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ria can be an optionally substituted alkyl, alkoxy, or -(CH2)o-7-NR3aR3b. In other embodiments, A can be selected from C3-Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)0-4-C(=Z)-ORi ; X can be S; Y can be -N(R2) =^= wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ri a can be an optionally substituted alkyl, alkoxy, or -(CH2)0-7-NR3aR3b. In still other embodiments, A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine); X can be S; Y can be -N(R2) = wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRuRib, wherein Ru can be an optionally substituted alkyl, alkoxy, or -(CH2)0-7-NR3aR3b. In yet still other embodiments, A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRuRib group; X can be S; Y can be -N(R2) ^^ wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ria can be an optionally substituted alkyl, alkoxy, or -(CH2)o.7-NR3aR3b- In an embodiment, the alkyl can be Ci-6 alkyl. In certain other embodiments, the alkoxy is a Ci-6 alkoxy.
[0033] In some embodiments, A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)0-4- C(=Z)-ORi; X can be S; Y can be -N(R2) ^= wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In other embodiments, A can be selected from C3-Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)0-4-C(=Z)-ORi ; X can be S; Y can be -N(R2) = wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be
Figure imgf000018_0001
wherein Ri2 is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In still other embodiments, A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine); X can be S; Y can be -N(R2) = wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein R)3 is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In yet still other embodiments, A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or - NRiaRib group; X can be S; Y can be -N(R2) ^^ wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ru is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In an embodiment, the optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl is selected from:
Figure imgf000019_0001
of the embodiments, n can be 1 or 2.
[0034] In some embodiments, A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)0.4- C(=Z)-ORi; X can be S; Y can be -N(R2) =^= wherein the symbol ^-= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRit» wherein Rj3 is an optionally substituted aryl or aralkyl. In other embodiments, A can be selected from C3- Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)0-4-C(=Z)-ORi; X can be S; Y can be - N(R2) = wherein the symbol ="= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ru is an optionally substituted aryl or aralkyl. In still other embodiments, A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine); X can be S; Y can be -N(R2) ^^ wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted aryl or aralkyl. In yet still other embodiments, A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRiaRib group; X can be S; Y can be -N(R2) ^-= wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ru is an optionally substituted aryl or aralkyl. In an embodiment, the optionally
substituted aryl or aralkyl can be selected from:
Figure imgf000019_0002
Figure imgf000019_0003
,wherein Q can be -N(R4)-, oxygen or sulfur; and R4 can be hydrogen or Chalky!, and in some of the embodiments, n can be 1 or 2. [0035] In some embodiments, A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NR^Rib, and -(CH2)0.4- C(=Z)-ORi; X can be S; Y can be -N(R2) =^= wherein the symbol ^= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ri3 is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In other embodiments, A can be selected from C3-Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)o-4-C(=Z)-ORi; X can be S; Y can be -N(R2) ^^ wherein the symbol ^= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRit>, wherein Ri8 is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In still other embodiments, A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine); X can be S; Y can be -N(R2) =^÷ wherein the symbol ^= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ri3 is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In yet still other embodiments, A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or - NRiaRib group; X can be S; Y can be -N(R2) =^ wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ru is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In an embodiment, the optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl can be selected from:
Figure imgf000020_0001
[0036] In some embodiments, A can be selected from C3-Ci2alkyl, C4- Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)O-4- C(=Z)-ORi; X can be S; Y can be -N(R2) ^^ wherein the symbol = represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein R)3 is an optionally substituted heteroaryl or heteroaralkyl. In other embodiments, A can be selected from C3-Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)0-4-C(=Z)-ORi; X can be S; Y can be -N(R2) ^= wherein the symbol ^="= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted heteroaryl or heteroaralkyl. In still other embodiments, A can be selected from aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine); X can be S; Y can be -N(R2) = wherein the symbol ^^ represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted heteroaryl or heteroaralkyl. In yet still other embodiments, A can be selected from cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRiaRib group; X can be S; Y can be -N(R2) = wherein the symbol ^-= represents a double bond and R2 does not exist; and B can be -C(=Z)NRiaRib, wherein Ri3 is an optionally substituted heteroaryl or heteroaralkyl. In an embodiment, the optionally substituted heteroaralkyl is from the
group consisting of:
Figure imgf000021_0001
Figure imgf000021_0003
, and
Figure imgf000021_0002
, wherein Q can be oxygen or sulfur, and in some of the embodiments, n can be 1 or 2. In certain other embodiments,
the optionally substituted heteroaralkyl can
Figure imgf000021_0004
,wherein Q can be oxygen or sulfur, and in some of the embodiments, n can be 1 or 2. [0037] Some embodiments disclosed herein describe a compound of Formula (I) wherein X can be S; and A can be selected from a mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl and alkynyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl and heteroaralkyl. In an embodiment, A can be selected from a mono-substituted, poly- substituted, or unsubstituted, straight or branched alkyl; and mono-substituted, poly- substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl. In an embodiment, A can be a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; or mono-substituted, poly-substituted or unsubstituted aryl. In an embodiment, A can be a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; or mono-substituted, poly-substituted or unsubstituted aryl; X can be S; and -N(R2) =^=, where ^= is a double bond; and R2 is absent. In some embodiments, B can be -C(=Z)NRiaRib. In other embodiments, B can be -C(=Z)Ri. In still other embodiments, B can be -S(O)2NRiaRib- In an embodiment, when B is -C(=Z)NRiaRib or -C(=Z)Ri, Z can be O (oxygen). In some embodiments, Ri1 Ri3 and Rib can be independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl. In an embodiment, Ri and Ria can be independently selected from alkyl, cycloalkyl, heteroaryl, heteroalicyclyl and heteroaralkyl; and R^ is hydrogen. In some embodiments, Y can be -N(R2) =-^, where = is a double bond; and R2 is absent. In an embodiment, A can be selected from a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl; B can be -C(=Z)NR]aRib; Ria can be independently selected from alkyl, cycloalkyl, heteroalicyclyl and heteroaralkyl; Rib is hydrogen; Y can be -N(R2) =^=, where ^= is a double bond; and R2 is absent. In an embodiment, A can be selected from a mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl; B can be -C(=Z)NRiaRib; Ria can be independently selected from alkyl, cycloalkyl, heteroalicyclyl and heteroaralkyl; R^ is hydrogen; Y can be -N(R2) ^=, where =^-= is a double bond; and R2 is absent. In still another embodiment, A can be selected from a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl; B can be -C(=Z)Ri; Ri can be alkyl; Y can be -N(R2) ^^, where === is a double bond; and R2 is absent. In yet still another embodiment, A can be selected from a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl; B can be -S(O)2NRi aRit,; Ru can be alkyl or heteroalicyclyl; Rib is hydrogen; Y can be -N(R2) ^^, where =^ is a double bond; and R2 is absent.
[0038] In some embodiments, Ari is phenyl. In other embodiments, Ari is selected from optionally substituted pyrazine, optionally substituted pyridine, optionally substituted pyrimidine, optionally substituted oxazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted pyrrole, optionally substituted imidazole, and optionally substituted phenyl. In some embodiments, Ar2 is selected from optionally substituted pyrazine, optionally substituted pyridine, optionally substituted indole, optionally substituted pyrimidine, optionally substituted oxazole, optionally substituted thiazole, optionally substituted furan, optionally substituted thiophene, optionally substituted pyrrole, optionally substituted imidazole, optionally substituted triazaole, optionally substituted isoxazole, optionally substituted isothiazole, optionally substituted pyrazole, and optionally substituted phenyl.
[0039] In an embodiment, the compound of Formula (I) can further include a detectable label such as a radiolabel. Exemplary detectable labels include, but are not limited to [3H], [18F], [11C] and [125I].
[0040] In various embodiments, compounds according to Formula (I) include the following structures:
Figure imgf000024_0001
22 23 24
Figure imgf000025_0002
Figure imgf000026_0001
40 41 42
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
82 83 84
Figure imgf000029_0002
Figure imgf000030_0001
-29-
Figure imgf000031_0001
115
[0041] Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of X can be combined with any one or more embodiments of Y, A, Ar1, Ar2, D, B, R1, R1,, R,b, R2, R3, R-3a, and R3b.
[0042] Other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Y can be combined with any one or more embodiments of X, A, Ar1, Ar2, D, B, R1, Rla, Rlb, R2, R3, Raa, and R3b.
[0043] Still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ari can be combined with any one or more embodiments of X, Y, A, Ar2, D, B, R1, Rla, Rib, R2, R3, R3a, and R3b. [0044] Yet still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ar2 can be combined with any one or more embodiments of X, Y, A, An, D, B, Ri, Ri3, Rn,, R2, R3, R3a and R3b.
[0045] Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of D can be combined with any one or more embodiments of X, Y, A, An, Ar2, B, R1, Rla, Rn,, R2, R3, R-3a and R3b.
[0046] Other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of B can be combined with any one or more embodiments of X, Y, A, Ar1, Ar2, D, R1, Rla, Rlb, R2, R3, R3a and R3b.
[0047] Still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Ri can be combined with any one or more embodiments of X, Y, A, Ari, Ar2, B, D, Ria, Rn,, R2, R3, R3a and R3b.
[0048] Yet still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R)a can be combined with any one or more embodiments of X, Y, A, Ari, Ar2, B, D, Ri, Rn,, R2, R3, R3a and R3b.
[0049] Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of Rib can be combined with any one or more embodiments of X, Y, A, Ar,, Ar2, B, D, R1, Rla, R2, R3, R3a and R3b.
[0050] Other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R2 can be combined with any one or more embodiments of X, Y, A, Ar1, Ar2, B, D, R1, Rla, Rib, R3, R3a and R3b.
[0051] Still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R3 can be combined with any one or more embodiments of X, Y, A, Ar1, Ar2, B, D, Rt, Ri3, Rib, R2, R3a and R3b.
[0052] Yet still other embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R3a can be combined with any one or more embodiments of X, Y, A, An, Ar2, B, D, Ri, Ria, Rib, R2, R3 and R3b.
[0053] Some embodiments disclosed herein relate to a compound of Formula (I), in which any embodiment of R3b can be combined with any one or more embodiments of X, Y, A, Ar1, Ar2, B, D, R1, R,a, R,b, R2, R3 and R3a. [0054] Certain of the compounds of the present invention may exist as stereoisomers including optical isomers. The invention includes all stereoisomers and both the racemic mixtures of such stereoisomers as well as the individual enantiomers that may be separated according to methods that are well known to those of ordinary skill in the art.
[0055] In some embodiments, the compound of Formula (I) can bind to a cannabinoid receptor. Preferably, in some embodiments, the cannabinoid receptor can be a CBl receptor.
Definitions
[0056] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. All patent, applications, published applications and other publications referenced herein are incorporated by reference in their entirety. In the event that there are plurality of definitions for a term herein, those in this section prevail unless stated otherwise.
[0057] As used herein, any "R" group(s) such as, without limitation, Ri, R!a and Rib, represent substituents that can be attached to the indicated atom. A non-limiting list of R groups include but are not limited to hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl. An R group may be substituted or unsubstituted. If two "R" groups are covalently bonded to the same atom or to adjacent atoms, then they may be "taken together" as defined herein to form a cycloalkyl, aryl, heteroaryl or heteroalicyclyl group. For example, without limitation, if Ra and Rb of an NR3Rb group are indicated to be "taken together", it means that they are covalently bonded to one another at their terminal atoms to form a ring that includes the nitrogen:
Figure imgf000033_0001
[0058] As used herein, "IC50" refers to an amount, concentration, or dosage of a particular test compound that achieves a 50% inhibition of a maximal response, such as modulation of GPCR, including cannabinoid receptor, activity an assay that measures such response. The assay may be an R-SAT® assay as described herein but is not limited to an RSAT assay.
[0059] As used herein, "EC50" refers to an amount, concentration or dosage of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound, in an assay that measures such response such as but not limited to R-SAT® assay described herein.
[0060] Whenever a group of this invention is described as being "optionally substituted" that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, when a group is described as being "unsubstituted or substituted" if substituted, the substituent may be selected from one or more the indicated substituents.
[0061] Unless otherwise indicated, when a substituent is deemed to be "optionally substituted," or "substituted" it is meant that the substituent is a group that may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. The protecting groups that may form the protective derivatives of the above substituents are known to those of skill in the art and may be found in references Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, which is hereby incorporated by reference in its entirety.
[0062] As used herein, "Cm to Cn" in which "m" and "n" are integers refers to the number of carbon atoms in an alkyl, alkenyl or alkynyl group or the number of carbon atoms in the ring of a cycloalkyl or cycloalkenyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl or ring of the cycloalkenyl can contain from "m" to "n", inclusive, carbon atoms. Thus, for example, a "Ci to C4 alkyl" group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3-, CH3CH2-, CH3CH2CH2-, (CH3)2CH-, CH3CH2CH2CH2-, CH3CH2CH(CH3)- and (CH3)3C-. If no "m" and "n" are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl or cycloalkenyl group, the broadest range described in these definitions is to be assumed.
[0063] As used herein, "alkyl" refers to a straight or branched hydrocarbon chain fully saturated (no double or triple bonds) hydrocarbon group. The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as "1 to 20" refers to each integer in the given range; e.g., "1 to 20 carbon atoms" means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term "alkyl" where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 5 carbon atoms. The alkyl group of the compounds may be designated as "CpC4 alkyl" or similar designations. By way of example only, "Ci-C4 alkyl" indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, and the like.
[0064] The alkyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is(are) one or more group(s) individually and independently selected from alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. Wherever a substituent is described as being "optionally substituted" that substitutent may be substituted with one of the above substituents unless otherwise indicated.
[0065] As used herein, "alkenyl" refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. An alkenyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
[0066] As used herein, "alkynyl" refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. An alkynyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
[0067] As used herein, "aryl" refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system that has a fully delocalized pi-electron system throughout all the rings. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group of this invention may be substituted or unsubstituted. When substituted, hydrogen atoms are replaced by substituent group(s) that is(are) one or more group(s) independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof, unless the substituent groups are otherwise indicated. [0068] As used herein, "heteroaryl" refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system throughout all the rings), one or two or more fused rings that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. Examples of heteroaryl rings include, but are not limited to, furan, thiophene, phthalazine, pyrrole, oxazole, thiazole, imidazole, pyrazole, isoxazole, isothiazole, triazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine and triazine. A heteroaryl group of this invention may be substituted or unsubstituted. When substituted, hydrogen atoms are replaced by substituent group(s) that is(are) one or more group(s) independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof.
[0069] An "aralkyl" is an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, substituted benzyl, 2- phenylethyl, 3-phenylpropyl, and naphtylalkyl.
[0070] A "heteroaralkyl" is heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylmethyl, 3- thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl, and their substituted as well as benzo-fused analogs.
[0071] "Lower alkylene groups" are straight-chained tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (-CH2-), ethylene (-CH2CH2-), propylene (- CH2CH2CH2-), and butylene (-(CH2)4-) groups. A lower alkylene group may be substituted or unsubstituted.
[0072] As used herein, "alkylidene" refers to a divalent group, such as =CR'R", which is attached to one carbon of another group, forming a double bond, Alkylidene groups include, but are not limited to, methylidene (=CH2) and ethylidene (=CHCH3). As used herein, "arylalkylidene" refers to an alkylidene group in which either R' and R" is an aryl group. An alkylidene group may be substituted or unsubstituted.
[0073] As used herein, "alkoxy" refers to the formula -OR wherein R is an alkyl is defined as above, e.g. methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, amoxy, tert-amoxy and the like. An alkoxy may be substituted or unsubstituted.
[0074] As used herein, "alkylthio" refers to the formula -SR wherein R is an alkyl is defined as above, e.g. methylmercapto, ethylmercapto, n-propylmercapto, 1- methylethylmercapto (isopropylmercapto), n-butylmercapto, iso-butylmercapto, sec- butylmercapto, tert-butylmercapto, and the like. An alkylthio may be substituted or unsubstituted.
[0075] As used herein, "aryloxy" and "arylthio" refers to RO- and RS-, in which R is an aryl, such as but not limited to phenyl. Both an aryloxy and arylthio may be substituted or unsubstituted.
[0076] As used herein, "acyl" refers to a hydrogen, alkyl, alkenyl, alkynyl, or aryl connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl, and acryl. An acyl may be substituted or unsubstituted. An acyl may be substituted or unsubstituted.
[0077] As used herein, "cycloalkyl" refers to a completely saturated (no double bonds) mono- or multi- cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro-connected fashion. Cycloalkyl groups of this invention may range from C3 to Ci0, in other embodiments it may range from C3 to C6. A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. If substituted, the substituent(s) may be an alkyl or selected from those indicated above with regard to substitution of an alkyl group unless otherwise indicated.
[0078] As used herein, "cycloalkenyl" refers to a cycloalkyl group that contains one or more double bonds in the ring although, if there is more than one, they cannot form a fully delocalized pi-electron system in the ring (otherwise the group would be "aryl," as defined herein). When composed of two or more rings, the rings may be connected together in a fused, bridged or spiro-connected fashion. A cycloalkenyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be an alkyl or selected from the groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
[0079] As used herein, "cycloalkynyl" refers to a cycloalkyl group that contains one or more triple bonds in the ring. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro-connected fashion. A cycloalkynyl group of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be an alkyl or selected from the groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
[0080] As used herein, "heteroalicyclic" or "heteroalicyclyl" refers to a stable 3- to 18 membered ring which consists of carbon atoms and from one to five heteroatoms selected from nitrogen, oxygen and sulfur. For the purpose of this invention, the "heteroalicyclic" or "heteroalicyclyl" may be monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may be joined together in a fused, bridged or spiro-connected fashion; and the nitrogen, carbon and sulfur atoms in the "heteroalicyclic" or "heteroalicyclyl" may be optionally oxidized; the nitrogen may be optionally quatemized; and the rings may also contain one or more double bonds provided that they do not form a fully delocalized pi-electron system throughout all the rings. Heteroalicyclyl groups of this invention may be unsubstituted or substituted. When substituted, the substituent(s) may be one or more groups independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. Examples of such "heteroalicyclic" or "heteroalicyclyl" include but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, moφholinyl, oxiranyl, piperidinyl N-Oxide, piperidinyl, piperazinyl, pyrrolidinyl, 4-piperidonyl, pyrazolidinyl, 2-oxopyrrolidinyl, thiamoφholinyl, thiamorpholinyl sulfoxide, and thiamoφholinyl sulfone unless the substituent groups are otherwise indicated.
[0081] A "(heteroalicyclyl)alkyl" is a heterocyclic or a heterocyclyl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclic or a heterocyclyl of a (heteroalicyclyl)alkyl may be substituted or unsubstituted. Examples include but are not limited 4-methyltetrahydro-2H-pyran, substituted 4-methyltetrahydro-2H-pyran, 4-ethylpiperidine, 4-propylpiperidine, 4- methyltetrahydro-2H-thiopyran, and 4-methyl-l,3-thiazinane.
[0082] A "(cycloalkyl)alkyl" is a cycloalkyl group connected, as a substituent, via a lower alkylene group. The lower alkylene and cycloalkyl of a (cycloalkyl)alkyl may be substituted or unsubstituted. Examples include but are not limited cyclopropylmethyl, cyclobutylmethyl, cyclopropylethyl, cyclopropylbutyl, cyclobutylethyl, cyclopropylisopropyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, cycloheptylmethyl, and the like.
[0083] A "(cycloalkenyl)alkyl" is a cycloalkenyl group connected, as a substituent, via a lower alkylene group. The lower alkylene and cycloalkenyl of a (cycloalkenyl)alkyl may be substituted or unsubstituted.
[0084] A "(cycloalkynyl)alkyl" is a cycloalkynyl group connected, as a substituent, via a lower alkylene group. The lower alkylene and cycloalkynyl of a (cycloalkynyl)alkyl may be substituted or unsubstituted.
[0085] As used herein, "halo" or "halogen" refers to F (fluoro), Cl (chloro), Br (bromo) or I (iodo). [0086] As used herein, "haloalkyl" refers to an alkyl group in which one or more of the hydrogen atoms are replaced by halogen. Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifiuoromethyl and l-chloro-2- fluoromethyl, 2-fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.
[0087] As used herein, "haloalkoxy" refers to RO-group in which R is a haloalkyl group. Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy and l-chloro-2-fluoromethoxy, 2- fluoroisobutyoxy. A haloalkoxy may be substituted or unsubstituted.
[0088] An "O-carboxy" group refers to a "RC(=O)O-" group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein. An O- carboxy may be substituted or unsubstituted.
[0089] A "C-carboxy" group refers to a "-C(O)R" group in which R can be the same as defined with respect to O-carboxy. A C-carboxy may be substituted or unsubstituted.
[0090] A "trihalomethanesulfonyl" group refers to an "X3CSO2-" group wherein X is a halogen.
[0091] A "cyano" group refers to a "-CN" group.
[0092] An "isocyanato" group refers to a "-NCO" group.
[0093] A "thiocyanato" group refers to a "-CNS" group.
[0094] An "isothiocyanato" group refers to an " -NCS" group.
[0095] A "sulfinyl" group refers to an "-S(O)-R" group in which R can be the same as defined with respect to O-carboxy. A sulfinyl may be substituted or unsubstituted.
[0096] A "sulfonyl" group refers to an "SO2R" group in which R can be the same as defined with respect to O-carboxy. A sulfonyl may be substituted or unsubstituted.
[0097] An "S-sulfonamido" group refers to a "-SO2NRARB" group in which RA and RB can be the same as R defined with respect to O-carboxy. An S-sulfonamido may be substituted or unsubstituted. [0098] An "N-sulfonamido" group refers to a "RSO2N(RA)-" group in which R and RA can be the same as R defined with respect to O-carboxy. A sulfonyl may be substituted or unsubstituted.
[0099] A "trihalomethanesulfonamido" group refers to an "X3CSO2N(R)-" group with X as halogen and R can be the same as defined with respect to O-carboxy. A trihalomethanesulfonamido may be substituted or unsubstituted.
[0100] An "O-carbamyl" group refers to a "-OC(=O)NRARB" group in which RA and RB can be the same as R defined with respect to O-carboxy. An O-carbamyl may be substituted or unsubstituted.
[0101] An "N-carbamyl" group refers to an "ROC(=O)NRA -" group in which R and RA can be the same as R defined with respect to O-carboxy. An N-carbamyl may be substituted or unsubstituted.
[0102] An "O-thiocarbamyl" group refers to a "-OC(=S)-NRARB" group in which RA and RB can be the same as R defined with respect to O-carboxy. An O-thiocarbamyl may be substituted or unsubstituted.
[0103] An "N-thiocarbamyl" group refers to an "ROC(=S)NRA-" group in which R and RA can be the same as R defined with respect to O-carboxy. An N-thiocarbamyl may be substituted or unsubstituted.
[0104] A "C-amido" group refers to a "-C(=0)NRARB" group in which RA and RB can be the same as R defined with respect to O-carboxy. A C-amido may be substituted or unsubstituted.
[0105] An "N-amido" group refers to a "RC(=O)NRA-" group in which R and RA can be the same as R defined with respect to O-carboxy. An N-amido may be substituted or unsubstituted.
[0106] An "ester" refers to a "-C(=O)OR" group in which R can be the same as defined with respect to O-carboxy. An ester may be substituted or unsubstituted.
[0107] A lower aminoalkyl refers to an amino group connected via a lower alkylene group. A lower aminoalkyl may be substituted or unsubstituted.
[0108] A lower alkoxyalkyl refers to an alkoxy group connected via a lower alkylene group. A lower alkoxyalkyl may be substituted or unsubstituted. [0109] Any unsubstituted or monosubstituted amine group on a compound herein can be converted to an amide, any hydroxyl group can be converted to an ester and any carboxyl group can be converted to either an amide or ester using techniques well- known to those skilled in the art (see, for example, Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999).
[0110] Where the numbers of substiruents are not specified (e.g. haloalkyl), there may be one or more substituents present. For example "haloalkyl" may include one or more of the same or different halogens. As another example, "Ci-C3 alkoxyphenyl" may include one or more of the same or different alkoxygroups containing one, two or three atoms.
[0111] As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (See, Biochem. 11:942-944 (1972)).
[0112] As employed herein, the following terms have their accepted meaning in the chemical literature.
AcOH acetic acid anhyd anhydrous aq aqueous
CDI 1 , 1 '-carbonyldiimidazole
DCM dichloromethane
DMF N,N-dimethylformamide
DMSO dimethyl sulfoxide
Et2O diethyl ether
EtOAc ethyl acetate
EtOH ethanol
MeOH methanol
NH4OAc ammonium acetate
Pd/C palladium on activated carbon r.t. room temperature [0113] It is understood that, in any compound of this invention having one or more chiral centers, if an absolute stereochemistry is not expressly indicated, then each center may independently be of R-configuration or S -configuration or a mixture thereof. Thus, the compounds provided herein may be enatiomerically pure or be stereoisomeric mixtures. In addition it is understood that, in any compound of this invention having one or more double bond(s) generating geometrical isomers that can be defined as E or Z each double bond may independently be E or Z a mixture thereof. Likewise, all tautomeric forms are also intended to be included.
[0114] As used herein, "pharmaceutically acceptable salt" refers to a salt of a compound that does not abrogate the biological activity and properties of the compound. Pharmaceutical salts can be obtained by reaction of a compound disclosed herein with an acid or base. Base-formed salts include, without limitation, ammonium salt (NH4 +); alkali metal, such as, without limitation, sodium or potassium, salts; alkaline earth, such as, without limitation, calcium or magnesium, salts; salts of organic bases such as, without limitation, dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine; and salts with the amino group of amino acids such as, without limitation, arginine and lysine. Useful acid-based salts include, without limitation, hydrochlorides, hydrobromides, sulfates, nitrates, phosphates, methanesulfonates, ethanesulfonates, p-toluenesulfonates and salicylates.
[0115] Pharmaceutically acceptable solvates and hydrates are complexes of a compound with one or more solvent of water molecules, or 1 to about 100, or 1 to about 10, or one to about 2, 3 or 4, solvent or water molecules.
[0116] As used herein, a "prodrug" refers to a compound that may not be pharmaceutically active but that is converted into an active drug upon in vivo administration. The prodrug may be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. Prodrugs are often useful because they may be easier to administer than the parent drug. They may, for example, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have better solubility than the active parent drug in pharmaceutical compositions. An example, without limitation, of a prodrug would be a compound disclosed herein, which is administered as an ester (the "prodrug") to facilitate absorption through a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to a carboxylic acid (the active entity) once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized in vivo to release the active parent compound. By virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, those skilled in the art, once a pharmaceutically active compound is known, can design prodrugs of the compound (see, e.g. Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392)
[0117] As used herein, the term "complement" refers to a oligonucleotide or polynucleotide that hybridizes by base-pairing, adenine to tyrosine and guanine to cytosine, to another oligonucleotide.
[0118] As used herein, to "modulate" the activity of CBl means either to activate it, i.e., to increase its cellular function over the base level measured in the particular environment in which it is found, or deactivate it, i.e., decrease its cellular function to less than the measured base level in the environment in which it is found and/or render it unable to perform its cellular function at all, even in the presence of a natural binding partner. A natural binding partner is an endogenous molecule that is an agonist for the receptor.
[0119] As used herein, to "detect" changes in the activity of CBl or of a CBl sub-type refers to the process of analyzing the result of an experiment using whatever analytical techniques are best suited to the particular situation. In some cases simple visual observation may suffice, in other cases the use of a microscope, visual or UV light analyzer or specific protein assays may be required. The proper selection of analytical tools and techniques to detect changes in the activity of CBl or a CBl sub-type are well- known to those skilled in the art.
[0120] An "agonist" is defined as a compound that increases the basal activity of a receptor (i.e. signal transduction mediated by the receptor). [0121] As used herein, "partial agonist" refers to a compound that has an affinity for a receptor but, unlike an agonist, when bound to the receptor it elicits only a fractional degree of the pharmacological response normally associated with the receptor even if a large number of receptors are occupied by the compound.
[0122] An "inverse agonist" is defined as a compound, which reduces, or suppresses the basal activity of a receptor, such that the compound is not technically an antagonist but, rather, is an agonist with negative intrinsic activity.
[0123] As used herein, "antagonist" refers to a compound that binds to a receptor to form a complex that does not give rise to any response, as if the receptor were unoccupied. An antagonist attenuates the action of an agonist on a receptor. An antagonist may bind reversibly or irreversibly, effectively eliminating the activity of the receptor permanently or at least until the antagonist is metabolized or dissociates or is otherwise removed by a physical or biological process.
[0124] As used herein, a "subject" refers to an animal that is the object of treatment, observation or experiment. "Animal" includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals. "Mammal" includes, without limitation, mice; rats; rabbits; guinea pigs; dogs; cats; sheep; goats; cows; horses; primates, such as monkeys, chimpanzees, and apes, and, in particular, humans.
[0125] As used herein, a "patient" refers to a subject that is being treated in order to attempt to cure, or at least ameliorate the effects of, a particular disease or disorder or to prevent the disease or disorder from occurring in the first place.
[0126] As used herein, the terms "treating," "treatment," "therapeutic," or "therapy" do not necessarily mean total cure or abolition of the disease or condition. Any alleviation of any undesired signs or symptoms of a disease or condition, to any extent can be considered treatment or therapy. Furthermore, treatment may include acts that may worsen the patient's overall feeling of well-being or appearance.
[0127] As used herein, a "carrier" refers to a compound that facilitates the incorporation of a compound into cells or tissues. For example, without limitation, dimethyl sulfoxide (DMSO) is a commonly utilized carrier that facilitates the uptake of many organic compounds into cells or tissues of a subject.
[0128] As used herein, a "diluent" refers to an ingredient in a pharmaceutical composition that lacks pharmacological activity but may be pharmaceutically necessary or desirable. For example, a diluent may be used to increase the bulk of a potent drug whose mass is too small for manufacture or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation. A common form of diluent in the art is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mimics the composition of human blood.
[0129] As used herein, an "excipient" refers to an inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition. A "diluent" is a type of excipient.
Synthesis
[0130] General synthetic routes to the compounds of this invention are shown in Schemes 1-12. The routes shown are illustrative only and are not intended, nor are they to be construed, to limit the scope of this invention in any manner whatsoever. Those skilled in the art will be able to recognize modifications of the disclosed synthesis and to devise alternate routes based on the disclosures herein; all such modifications and alternate routes are within the scope of this invention.
Scheme 1
Figure imgf000048_0001
[0131] In Scheme 1, Ri3 and A are as defined above for Formula I. R4, R5, and R6 are appropriate radicals selected to obtain the desired A group. By appropriate selection of Ri3, R4, R5, and R6 compounds 2, 5, 6, 10, 16, 26, 28 - 31, 40, 44, 46, 48, 51, and 58 can be prepared according to Scheme 1. For example, compound 2 can be prepared by using /j-butyl amine as H2N-Ri3 and 2-pyridyl zinc bromide as R6ZnX. Suitable heterocyclic 2-mercaptosubstituted carboxylic acids can be prepared as described in Blank et al, J. Med. Chem. 1977, 20, 572-576; Sen et al, Indian J Chem B Org, 1981, 2OB, 275-278, and Solomon et al, Heterocycles, 1987, 26, 651-674), all of which are incorporated herein by reference in their entirety.
Scheme 2
Figure imgf000048_0002
[0132] In Scheme 2, Ri3 and A are as defined above for Formula I. R4, R5, and R6 are appropriate radicals selected to obtain the desired A group. By appropriate selection of Ri3, R4, R5, and R6 compounds 13-16, 18, 36-42, 54, 55, 57-60 can be prepared according to Scheme 2. For example, compound 13 can be prepared by using cyclohexyl amine a H2N-Ru and p-Cl-phenyl zinc iodide as R6ZnX. Other suitable heterocyclic 2-chloro carboxylic acids include commercially available 2-chloro nicotinic acid, 3-chloro-4-pyridine carboxylic acid, 4-chloronicotinic acid and 3-chloro-2-pyrazine carboxylic acid and those synthesized by methods described in Bredereck et al. Chem. Ber, 1962, 95, 956-963 and Krasovskiy et al., Angew. Chem. Int. Ed., 2006, 45, 2958- 2961, both of which are incorporated herein by reference in their entirety.
Scheme 3
Figure imgf000049_0001
[0133] Scheme 3 illustrates one method of obtaining compound 7. By employing other optionally substituted iodo-or bromo benzoheterophenones followed by the appropriate amine in the amide coupling step, compounds 1, 3, 8, 9, 11, 19-25, 27, 32- 34, 43, 45, 49, 50, 52, 53, 61, 62, 82-84 can be obtained using a similar method. Suitable iodo-or bromo benzoheterophenones can be obtained commercially, synthesized by the methods described in Reynolds et al, Tetrahedron, 2001, 57, 7765-7770, Liu et al., Org. Lett. 2006, 8, 617-619 or by generation of zinc reagents from dibromo- or diiodo heterocycles followed by reaction with the appropriately substituted benzoyl chloride (Knochel et al., Angew. Chem. 2006). All of the foregoing references are incorporated herein by reference in their entirety.
Scheme 4
Figure imgf000050_0001
COI SOCI2/cat DMF 3 NHR4R5
THF
Figure imgf000050_0002
T Tooluueennee
Figure imgf000050_0004
Figure imgf000050_0003
Figure imgf000050_0005
[0134] In Scheme 4, Ria and A are as defined above for Formula I. R4, R5, and R6 are appropriate radicals selected to obtain the desired A group. By appropriate selection of Ri3, R4, R5, and R6 compounds 4, 12, 17, 47, 56, 63, and 84 can be prepared according to Scheme 4 by using the appropriate heterocyclic 2-mercaptosubstituted carboxylic acids followed by the appropriate alkyl or aryl metal halide or amine and the desired amine in the last step.
Scheme 5
Figure imgf000050_0006
[0135] In Scheme 5, Ria and A are as defined above. R4, R5, and R6 are appropriate radicals selected to obtain the desired A group. By appropriate selection of Ria, R4, R5, and R6 compounds 64, 68, 72, 78, 88, 92, and 96 can be prepared by using the appropriate heterocycle, amine, metal reagent/amine and amine. Further relevant synthetic methods can be found in Thompson et al, J. Org. Chem. 1988, 53, 2052-2055, which is incorporated herein by reference in its entirety.
Scheme 6
Figure imgf000051_0001
[0136] In Scheme 6, Ru and A are as defined above for Formula I. R4, R5, and R6 are appropriate radicals selected to obtain the desired A group. By appropriate selection of Ru, R4, R5, and R6 compounds 64-74, 85, 86, 88-91, 93- 95, 97 -101, and 104 can be prepared by using appropriate pyridines obtained either commercially or synthesized by magnesization of heterocycles via selective deprotonation as described in Liu et al, Org. Lett. 2006, 8, 617-619, incorporated herein by reference in its entirety, combined with the above mentioned 2-mercaptosubstituted carboxylic acids/esters followed by the appropriate reagents in the reaction sequence. In addition, compounds 80, 81, 103, and 105 can be prepared according to Scheme 6 using pyrimidines obtained as described in Krasovskiy et al, Angew. Chem. Int. Ed. 2006, 45, 2958-2961, which is incorporated herein by reference in its entirety. Scheme 7
Figure imgf000052_0001
[0137] Scheme 7 illustrates one method of obtaining compound 102. Other pyrazines such as compounds 87 and 102 can be obtained in a similar fashion following the methods described in PIe et al., J. Org. Chem. 1995, 60, 3781-3786, which is incorporated herein by reference in its entirety. Pyrazines containing an amide side chain instead of sulfonamide side chain can be obtained by reacting the metalated species with a variety of alkyl isocyanates.
Scheme 8 i. 2 equiv n-BuLi, 2 equiv TMEDA, cyclohexane ii. 1 equiv ACN
Figure imgf000052_0003
Figure imgf000052_0002
[0138] In Scheme 8, Ari, Ar2, B, and D are as described above for Formula I. A is an aryl or heteroaryl. X is Br, Cl, F, OTf, or OTs. Scheme 8 proceeds by dilithiation of arylthiols or heteroaryl thiols as described in Figuly et al, J. Am. Chem. Soc, 1989, 111, 654, Block et al, J. Am. Chem. Soc, 1989, 111, 658, and Smith et al, J. Am. Chem. Soc, 1989, 111, 665, which are incorporated herein by reference in their entirety. Upon addition of aromatic or a heteroaromatic nitriles as described in Brieaddy and Donaldson, J. Heterocyclic Chem., 1995, 32, 1683 and Katritzky et al, Chem. Heterocyclic Compd. Eng. Trans., 2002, 38, 156, which are incorporated herein by reference in their entirety, the dilithioketimine intermediate is obtained. This intermediate can be in turn treated with suitable ø-dihaloaryls or o-dihaloheteroaryls to obtain the thiazepine product. Specifically, but not exclusively, π-deficient o-dihaloazaarenes constitute outstanding substrates for the reactions with ketimines, providing the thiazepine products in high yield simply by adding 1 equiv of a neat π-deficient ø-dihaloazaarene to a reaction mixture containing a premade ketimine.
Scheme 9
or MgCI
Figure imgf000053_0001
Figure imgf000053_0002
[0139] In Scheme 9, Ar2 and D are as described above for Formula I. A is an aryl or heteroaryl. X is Br, Cl, F, OTf, or OTs. Ari is a π-deficient azaaryl. E is an electrophile including, but not limited to, isocyanates, acid chlorides, nitriles, tosyl cyanide or 1-cyanoimidazole, aldehydes or ketones, halogens or organic halides, carbon dioxide, Weinreb amides, tosyl azide, zinc chloride, tin chloride and trimethyl borate.
[0140] On the left side of Scheme 9, an electrophile is added to a π-deficient- azaaryl-containing thiazepines by means of α-lithiation (Queguiner et al, Adv. Heterocycl. Chem. 1991, 52, 187, Gros et al, J. Org. Chem. 2002, 67, 234, Smith et al, Org. Lett.
2005, 7, 5457, incorporated by reference in its entirety), α-zincation (Kondo et al, J. Am. Chem. Soc. 1999, 121, 3539, Imahori et al, Chem. Commun. 2001, 2450, incorporated by reference in its entirety) or α-magnesiation (Krasovskiy et al, Angew. Chem Int. Ed.,
2006, 45, 2958, incorporated by reference in its entirety). The organometallic intermediates thus generated can be treated with convenient electrophiles to give substituted thiazepines. In the cases where such substitution tactics afford a mixture of two or more regioisomeric products, the products can be easily separated from each other by one of the standard methods known in the art.
[0141] On the right side of Scheme 9, an electrophile can be added to a π- defϊcient ø-dihaloazaryl by a similar method as described above. The resulting heteroaryl can be in turn be reacted with a ketimine to produce the desired thiazepine product. Convenient substituents that can be introduced on the azaryl include, but are not limited to, nitrile or ester moieties. Introduction of such electron withdrawing groups on the rings of azaryls improves their reactivity towards ketimines. Also, at a later stage, these groups can be easily converted to other pertinent functional groups.
Scheme 10
Figure imgf000054_0001
[0142] In Scheme 10, Ari, Ar2, B, and D are as described above for Formula I. A is an alkyl, aryl, heteroaryl, or amino. X is Br, Cl, F, OTf, or OTs. Scheme 10 provides alkyl and amino substituted thiazepines by addition of 2 equiv of alkyllithiums, aryllithiums, heteroaryllithiums or 2 equiv of lithium amides to 2-cyanoarene thiols, giving rise to a ketimine, which can be reacted with ø-dihaloaryl (or heteroaryl) to produce the desired thiazepine product. The requisite 2-cyanoaryl thiols can be prepared for example by heating a 2-cyanobromoaryl (or heteroaryl) with mercaptoacetic ethyl ester in the presence of potassium t-butoxide and liquid ammonia (Brugelmans et al, Tetrahedron, 1983, 39, 4153, which is incorporated herein by reference in its entirety).
Scheme 11
Figure imgf000054_0002
[0143] In Scheme 11, AIΪ, Ar2, B, and D are as described above for Formula I. A is an alkyl, amino, aryl, or heteroaryl. X is I, Br, or Cl. Scheme 11 provides thiazepines by hydrolysis of ketimines followed by reacting the resulting ketoaryl thiol with a suitable o-aminohaloaryl (or heteroaryl). The cyclization to produce the thiazepines can be carried under copper catalysis (Bates et al, Org. Lett., 2002, 4, 2803, Kwong et al, Org. Lett., 2002, 4, 3517, which is incorporated herein by reference in its entirety) using a ketone and ø-aminoiodoaryl (or heteroaryl); or under palladium catalysis (Li, J. Org. Chem., 2001, 66, 8677, Li, Angew. Chem Int. Ed., 2001, 40, 1513, which is incorporated herein by reference in its entirety) using a ketone and o-aminobromo(chloro or bromo)aryl (or heteroaryl). The ketones can also be prepared by other methods known in the art, for example by addition of organolithiums to mercaptoaryl(or heteroaryl)carboxylic acids (Bull. Chem. Soc. jpn. 2001, 77, 2095, which is incorporated herein by reference in its entirety) or by thermal rearrangement of thiocarbamate esters followed base hydrolysis (Tremont et al, J. Med. Chem., 2005, 18, 5837, which is incorporated herein by reference in its entirety).
Scheme 12
Figure imgf000055_0001
[0144] In Scheme 12, Ari, Ar2, B, and D are as described above for Formula I. A is an alkyl, amino, aryl, or heteroaryl. X is I, Br, or Cl. M is a main group metal. Scheme 12 described the production of thiazepines by reacting a haloketones with an o- aryl(or heteroaryl) thiol. In particular, such cyclizations can be carried out very efficiently under copper catalysis (Kwong et al, Org. Lett., 2002, 4, 3517, incorporated herein by reference in its entirety) using iodoketones and o-aminoaryl(or heteroaryl) thiols. The requisite iodoketones can be synthesized by one of the methods known in the art, for example by addition of organomagnesium reagents (Reynolds and Hermitage, Tetrahedron, 2001, 57, 7765, incorporated herein by reference in its entirety) to 2-iodo Weinreb amides (Brunette and Lipton, J. Org. Chem., 2000, 65, 5114, incorporated herein by reference in its entirety), by copper catalyzed aromatic Finkelstein reaction (Klapars and Buchwald, J. Am. Chem. Soc, 2002, 124, 14844, incorporated herein by reference in its entirety) using the more ubiquitous bromoketones (Krasovskiy and Knochel, Angew. Chem. 2004, 43, 3333, incorporated herein by reference in its entirety) or by diazotation/Sandmeyer-type iodination (J. Org. Chem., 1982, 47, 2441, incorporated herein by reference in its entirety) using the readily available aminoketones (Sternbach et al, J. Am. Chem. Soc, 1961, 26, 4488, incorporated herein by reference in its entirety). Likewise, the o-aminoaryl(or heteroaryl) thiol employed herein for the synthesis of thiazepines can be prepared by one of the methods known in the art, for example by reduction of ø-nitroaryl(or heteroaryl) thiols (Foster and Reid, J. Am. Chem. Soc, 1924, 46, 1936, incorporated herein by reference in its entirety), by heating of 2- halonitroaryls(or heteroaryls) with sodium sulphide in water (Jain et al, Chem. End., 1969, 989, incorporated herein by reference in its entirety) or by saponification of (het)arofused 2-substituted thiazoles (Hodson et al, J. Med. Chem., 2002, 45, 2229). Bromo(or chloro)ketones can also participate in reactions with o-aminoaryl(or heteroaryl) thiols using a palladium catalyst (Li, J. Org. Chem., 2002, 67, 3643, incorporated herein by reference in its entirety) to provide thiazepines.
[0145] Finally, under SNAΓ conditions, chloroketones can be reacted with o- aminoaryl(or heteroaryl) thiols to afford thiazepines. Presence of several strongly electron withdrawing groups (such as nitro or cyano groups) on the aromatic ring of the chloroketones (Jarret and Loudon, J. Chem. Soc, 1957, 3818, Gait and Loudon, J. Chem. Soc, 1959, 885, incorporated herein by reference in its entirety), or a π-deficient azaromatic chloroketone (Warmhof, Synthesis., 1972, 151, Shalaby, Phosphorus, Sulfur Relat. Elem., 2003, 775, 199, incorporated herein by reference in its entirety), is typically required.
Methods of Use
[0146] The term "therapeutically effective amount" is used to indicate an amount of an active compound, or pharmaceutical agent, that elicits the biological or medicinal response indicated. This response may occur in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, and includes alleviation of the symptoms of the disease being treated.
[0147] One embodiment disclosed herein relates to a method of ameliorating or treating a disease or condition by administering to a subject a therapeutically effective amount of one or more compounds of Formula I. The disease or condition can be selected from: obesity, metabolic syndrome, a metabolic disorder, hypertension, polycystic ovary disease, osteoarthritis, a deimatological disorder, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis, a sleep disorder, hyperlipidemic conditions, bulimia nervosa, a compulsive eating disorder, an appetite disorder, atherosclerosis, diabetes, high cholesterol, hyperlipidemia, cachexia, an inflammatory disease, rheumatoid arthritis, a neurological disorder, a psychiatric disorder, substance abuse (e.g., alcohol, amphetamines, amphetamine-like substances, caffeine, cannabis, ***e, hallucinogens, inhalents, nicotine, opioids, phencyclidine, phencyclidine-like compounds, sedative-hypnotics or benzodiazepines, and/or other unknown substances), depression, anxiety, mania, schizophrenia, dementia, dystonia, muscle spasticity, tremor, psychosis, an attention deficit disorder, a memory disorder, a cognitive disorder, short term memory loss, memory impairment (e.g., associated with dementia, Alzheimer's disease, schizophrenia, Parkinson's disease, Huntington's disease, Pick's disease, Creutzfeld- Jakob disease, HIV, cardiovascular disease, head trauma and/or age-related cognitive decline), drug addiction, alcohol addiction, nicotine addiction, infertility, hemorrhagic shock, septic shock, cirrhosis, a cardiovascular disorder, cardiac dysfunction, valvular disease, myocardial infarction, cardiac hypertrophy, congestive heart failure, transplant rejection, an intestinal disorder, a neurodegenerative disease, multiple sclerosis, Alzheimer's disease, Parkinson's disease, epilepsy, Huntington's disease, Tourette's syndrome, cerebral ischaemia, cerebral apoplexy, craniocerebral trauma, stroke, spinal cord injury, catabolism, hypotension, hemorrhagic hypotension, endotoxin-induced hypotension, an eye disorder, glaucoma, uveitis, retinopathy, dry eye, macular degeneration, emesis, nausea, a gastric ulcer, diarrhea, pain, a neuropathic pain disorder, viral encephalitis, plaque sclerosis, cancer, a bone disorder, bone density loss, osteoporosis, ostepenia, a lung disorder, asthma, pleurisy, polycystic ovary disease, premature abortion; inflammatory bowel disease, lupus, graft vs. host disease, T-cell mediated hypersensitivity disease, Hashimoto's thyroiditis, Guillain-Barre syndrome, contact dermatitis, allergic rhinitis, ischemic injury, and reperfusion injury. In one embodiment, the therapeutically effective amount of a compound of Formula (I) is in a sufficient amount to ameliorate or treat said disease or condition by binding to a cannabinoid receptor (e.g., CB-I receptor). In another embodiment, the method can further include identifying a subject in need of ameliorating or treating said disease or condition.
[0148] Also disclosed herein are methods of treating clinical manifestations in which a subject would benefit from modulation of the cannabinoid receptor (e.g., CB-I receptor), for example, antagonism of or inverse agonism of the cannabinoid receptor (e.g., CB-I receptor) wherein such modulation would treat clinical manifestations such as obesity, metabolic syndrome, a metabolic disorder, hypertension, polycystic ovary disease, osteoarthritis, a dermatological disorder, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis, a sleep disorder, hyperlipidemic conditions, bulimia nervosa, a compulsive eating disorder, an appetite disorder, atherosclerosis, diabetes, high cholesterol, hyperlipidemia, cachexia, an inflammatory disease, rheumatoid arthritis, a neurological disorder, a psychiatric disorder, substance abuse (e.g., alcohol, amphetamines, amphetamine-like substances, caffeine, cannabis, ***e, hallucinogens, inhalents, nicotine, opioids, phencyclidine, phencyclidine-like compounds, sedative-hypnotics or benzodiazepines, and/or other unknown substances), depression, anxiety, mania, schizophrenia, dementia, dystonia, muscle spasticity, tremor, psychosis, an attention deficit disorder, a memory disorder, a cognitive disorder, short term memory loss, memory impairment (e.g., associated with dementia, Alzheimer's disease, schizophrenia, Parkinson's disease, Huntington's disease, Pick's disease, Creutzfeld- Jakob disease, HIV, cardiovascular disease, head trauma and/or age-related cognitive decline), drug addiction, alcohol addiction, nicotine addiction, infertility, hemorrhagic shock, septic shock, cirrhosis, a cardiovascular disorder, cardiac dysfunction, valvular disease, myocardial infarction, cardiac hypertrophy, congestive heart failure, transplant rejection, an intestinal disorder, a neurodegenerative disease, multiple sclerosis, Alzheimer's disease, Parkinson's disease, epilepsy, Huntington's disease, Tourette's syndrome, cerebral ischaemia, cerebral apoplexy, craniocerebral trauma, stroke, spinal cord injury, catabolism, hypotension, hemorrhagic hypotension, endotoxin-induced hypotension, an eye disorder, glaucoma, uveitis, retinopathy, dry eye, macular degeneration, emesis, nausea, a gastric ulcer, diarrhea, pain, a neuropathic pain disorder, viral encephalitis, plaque sclerosis, cancer, a bone disorder, bone density loss, osteoporosis, ostepenia, a lung disorder, asthma, pleurisy, polycystic ovary disease, premature abortion; inflammatory bowel disease, lupus, graft vs. host disease, T-cell mediated hypersensitivity disease, Hashimoto's thyroiditis, Guillain-Barre syndrome, contact dermatitis, allergic rhinitis, ischemic injury, and reperfusion injury, comprising administering to a subject a therapeutically effective amount of a compound of Formula I. These methods include, but are not limited to methods such as a method of treating clinical manifestations in which cannabinoid receptor function is altered.
[0149] Some embodiments disclosed herein relate to a method for ameliorating or treating a disease or condition in which it would be beneficial to modulate the activity of a cannabinoid receptor, such as a CBl receptor, that can include administering to a subject a therapeutically effective amount of a compound of Formula I.
[0150] In an embodiment, the neurological disorder can be schizophrenia, dementia, dystonia, muscle spasticity, tremor, psychosis, anxiety, depression, an attention deficit disorder, a memory disorder, a cognitive disorder, drug addiction, alcohol addiction, nicotine addiction, a neurodegenerative disease, multiple sclerosis, Alzheimer's disease, Parkinson's disease, epilepsy, Huntington's disease, Tourette's syndrome, cerebral ischaemia, cerebral apoplexy, craniocerebral trauma, stroke, spinal cord injury, pain, neuropathic pain disorder, viral encephalitis, and/or plaque sclerosis.
[0151] In some embodiments, the disease or condition can be obesity, metabolic syndrome, appetite disorders, cachexia, high cholesterol, hyperlipidemia and/or diabetes.
[0152] In an embodiment, the disease or condition can be of the gastrointestinal system such as emesis, nausea, gastric ulcers, diarrhea and/or intestinal disorders.
[0153[ In some embodiments, the disease or disorder can be an inflammation disease (e.g., rheumatoid arthritis, asthma, psoriasis).
[0154] In an embodiment, the disease or condition can be of the cardiovascular system such as hemorrhagic sock, septic shock, cirrhosis, atherosclerosis, and/or cardiovascular disorders.
[0155] In other embodiments, the disease or condition can be of the reproductive system such as infertility and/or premature abortion.
[0156] In some embodiments, the disease or condition can be of the visual system such as glaucoma, uveitis, retinopathy, dry eye and/or macular degeneration.
[0157] In an embodiment, the disease or condition can be osteoporosis and/or ostepenia.
[0158] In other embodiments, the disease or condition can be asthma and/or pleurisy.
[0159] In an embodiment, the disease or condition can be cancer.
[0160] Another embodiment described herein relates to a method of ameliorating and/or treating drug and/or alcohol addiction comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
[0161] Still another embodiment described herein relates to a method for ameliorating or treating a disease or condition in which it would be beneficial to modulate the activity of a CBl receptor comprising administering to a subject a therapeutically effective amount of a compound of Formula (I). [0162] Yet still another embodiment described herein relates to a method of ameliorating and/or treating obesity, comprising administering to a subject a therapeutically effective amount of a compound of Formula (I).
[0163] One still another embodiment described herein relates to a method of ameliorating and/or treating impaired cognition and/or a memory disorder comprising administering to a subject a therapeutically effective amount of a compound of Formula
(I)-
[0164] Another embodiment described herein relates to a method of improving cognition or memory in a subject comprising administering to the subject a therapeutically effective amount of a compound of Formula (I)
[0165] Still another embodiment described herein relates to a method of ameliorating and/or treating inflammation due to an inflammatory disease comprising administering to a subject a therapeutically effective amount of a compound of Formula (I). A non-limiting list of inflammatory diseases include rheumatoid arthritis, asthma, and psoriasis.
[0166] Any of the embodiments listed herein may further include identifying a subject in need of treatment or amelioration of any disease or condition identified herein.
[0167] Other embodiments disclosed herein relate to a method of identifying a compound that treats or amerliorates any disease or condition identified herein in a subject, comprising identifying a subject suffering the disease or condition; providing the subject with at least one compound of Formula I, as defined herein; and determining if the at least one compound treats the disease or condition in the subject.
[0168] Some embodiments disclosed herein relate to a method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor in a subject that includes administering to the subject an effective amount of a compound of Formula I. In one embodiment, the cannabinoid receptor can be a CBl receptor.
[0169] Other embodiments disclosed herein relate to a method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor comprising contacting a cannabinoid receptor with a compound of Formula I. In one embodiment, the cannabinoid receptor can be a CBl receptor. [0170] Still other embodiments disclosed herein relate to a method of modulating or specifically inverse agonizing or antagonizing one or more cannabinoid receptors comprising identifying a subject in need of treatment or prevention and administering to the subject a therapeutically effective amount of a compound of Formula I.
[0171] Yet still other embodiments disclosed herein relate to a method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor that includes contacting a cannabinoid receptor with at least one test compound of Formula I; and determining any increase or decrease in activity level of the cannabinoid receptor so as to identify said test compound as an agonist, inverse agonist or antagonist of the cannabinoid receptor. In one embodiment, the cannabinoid receptor can be a CBl receptor. In another embodiment, the cannabinoid receptor can consists essentially of SEQ ID NO: 2. In yet still another embodiment, the cannabinoid receptor can have at least 90% amino acid identity to SEQ ED NO: 2. In one embodiment, the cannabinoid receptor can have at least 85% amino acid identity to SEQ ID NO: 2. In another embodiment, the cannabinoid receptor can have at least 70% amino acid identity to SEQ ED NO: 2.
[0172] One embodiment disclosed herein relates to a method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor that includes culturing cells that express a cannabinoid receptor; incubating the cells or a component extracted from the cells with at least one test compound of Formula I; and determining any increase or decrease in activity of the cannabinoid receptor so as to identify said test compound as an agonist, inverse agonist, or antagonist of the cannabinoid receptor. In one embodiment, the cannabinoid receptor can be a CBl receptor. In another embodiment, the cannabinoid receptor can consists essentially of SEQ ID NO: 2. In yet still another embodiment, the cannabinoid receptor can have at least 90% amino acid identity to SEQ ED NO: 2. In one embodiment, the cannabinoid receptor can have at least 85% amino acid identity to SEQ ED NO: 2. In another embodiment, the cannabinoid receptor can have at least 70% amino acid identity to SEQ ID NO: 2. [0173] Still another embodiment described herein relates to a method for identifying a compound which binds to a cannabinoid receptor comprising: labeling a compound described herein; with a detectable label; contacting the CB-I receptor with the labeled compound; and determining whether the labeled compound binds to the CB- 1 receptor. In an embodiment, the detectable label can be a radiolabel such as [3H], [18F], [11C] and [125IJ.
Pharmaceutical Compositions
[0174] In another aspect, the present invention relates to a pharmaceutical composition comprising a compound of Formula I as described above, and a physiologically acceptable carrier, diluent, or excipient, or a combination thereof.
[0175] The term "pharmaceutical composition" refers to a mixture of a compound disclosed herein with other chemical components, such as diluents or carriers. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, intramuscular, intraocular, intranasal, intravenous, injection, aerosol, parenteral, and topical administration. Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. Pharmaceutical compositions will generally be tailored to the specific intended route of administration.
[0176] The term "physiologically acceptable" defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
[0177] The pharmaceutical compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s). Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 18th edition, 1990, which is hereby incorporated by reference in its entirety. [0178] Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, intraocular injections or as an aerosol inhalant.
[0179] Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into the area of pain or inflammation, often in a depot or sustained release formulation. Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with a tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the organ.
[0180] The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.
[0181] Pharmaceutical compositions for use in accordance with the present disclosure thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., as disclosed in Remington's Pharmaceutical Sciences, cited above.
[0182] For injection, the agents disclosed herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
[0183] For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds disclosed herein to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with pharmaceutical combination disclosed herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[0184] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[0185] Pharmaceutical preparations, which can be used orally, include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
[0186] For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. [0187] For administration by inhalation, the compounds for use according to the present disclosure are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[0188] The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
[0189] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
[0190] Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[0191] The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
[0192] In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[0193] An exemplary pharmaceutical carrier for the hydrophobic compounds disclosed herein is a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. A common co-solvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80™, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of Polysorbate 80™; the fraction size of polyethylene glycol may be varied; and other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone. Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
[0194] Many of the compounds used in the pharmaceutical combinations disclosed herein may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free acids or base forms.
[0195] Pharmaceutical compositions suitable for use in the methods disclosed herein include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
[0196] The exact formulation, route of administration and dosage for the pharmaceutical compositions disclosed herein can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al. 1975, in "The Pharmacological Basis of Therapeutics", Chapter 1, which is hereby incorporated by reference in its entirety). Typically, the dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient's body weight, or 1 to 500 mg/kg, or 10 to 500 mg/kg, or 50 to 100 mg/kg of the patient's body weight. The dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient. Where no human dosage is established, a suitable human dosage can be inferred from ED50 or ID5o values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.
[0197] Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. The daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg and 500 mg of each ingredient, preferably between 1 mg and 250 mg, e.g. 5 to 200 mg or an intravenous, subcutaneous, or intramuscular dose of each ingredient between 0.01 mg and 100 mg, preferably between 0.1 mg and 60 mg, e.g. 1 to 40 mg of each ingredient of the pharmaceutical compositions disclosed herein or a pharmaceutically acceptable salt thereof calculated as the free base, the composition being administered 1 to 4 times per day. Alternatively the compositions disclosed herein may be administered by continuous intravenous infusion, preferably at a dose of each ingredient up to 400 mg per day. Thus, the total daily dosage by oral administration of each ingredient will typically be in the range 1 to 2000 mg and the total daily dosage by parenteral administration will typically be in the range 0.1 to 400 mg. In some embodiments, the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years.
[0198] Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety, which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
[0199] Dosage intervals can also be determined using MEC value. Compositions should be administered using a regimen, which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
[0200] In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
[0201] The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
[0202] The compositions may, if desired, be presented in a pack or dispenser device, which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. Compositions comprising a compound disclosed herein formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
[0203] It will be understood by those of skill in the art that numerous and various modifications can be made without departing from the spirit of the present disclosure. Therefore, it should be clearly understood that the forms disclosed herein are illustrative only and are not intended to limit the scope of the present disclosure.
EXAMPLES
[0204] Embodiments of the present invention are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the invention.
Example 1 : 10-Phenyl-5-thia- 1 ,4, 11 -triazadibenzo[a,dlcycloheptene
Figure imgf000070_0001
[0205] Thiophenol (5.0 g, 45.0 mmol) was added dropwise to «-BuLi in hexanes (42.8 mmol, 90 mmol) and tetramethylenediamine (13.6 mL, 90 mmol) in 4A mol sieves dried cyclohexane (50 mL), under Argon atmosphere at 0 0C. The resulting mixture was allowed to stir to r.t. overnight, then neat benzonitrile (4.59 mL, 45 mmol) was added dropwise at rt and the resulting brown suspension was stirred vigorously for 3 hours before adding neat 2,3-dichloropyrazine (6.8 g, 45 mmol) over ca. 1 min, causing a substantially exothermic reaction. After the heat evolution had ceased the crude reaction mixture was poured into 2M HCl (500 mL), extracted with toluene (3 x 250 mL), evaporated to dryness and passed through a pad of silica using EtOAc/«-heptane (1 : 2) as eluent to give 5.2 g (40 %) of the title compound as a brown solid.
[0206] LCMS m/z 290 [M+H]+. GCMS m/z 289 [M]+. 1H NMR (CDCl3) δ 7.28-7.56 (m, 6H), 7.67 (m, IH), 7.88 (m, 2H), 8.24 (d, 1H, J = 2.3), 8.41 (d, IH, J = 2.3). HPLC tR = 3.9 min (method B). GC tR = 9.7 min. Example 2: 3-Hvdroxy-2-methoxycarbonylthiophene
Figure imgf000071_0001
[0207] The title compound was synthesized according to a procedure published by Huddleston and Barker, Synth. Commun., 1979, 9, 8, 731-734, which is hereby incorporated by reference in its entirety. To a solution of 15 mL dry MeOH was added sodium (700 mg; 30 mmol) to give a 2M solution of NaOMe. Methyl thioglycolate (1.9 g; 18 mmol) was added. The solution was cooled to O0C and methyl 2-chloroacrylate (2.1 g; 17.4 mmol) was added drop wise. Stirring was continued overnight raising the temperature to room temperature. The mixture was re-cooled to 0°C and the quenched by addition of HCl (4M aq, ~5mL). Water was added and extraction done with 2 times EtOAc. The combined organic phases were dried over Na2SO4, filtered and the solvent removed in vacuo to give 2.0 g (70 %) of a brown oil that solidified upon drying. Used without further purification.
[0208] LCMS m/z -, tR = 4.12 min, purity (UV/MS) 98/20. GCMS m/z 158 (M), *R = 4.52 min. 1H NMR (CDCl3, 400 MHz) δ 9.56 (br s, IH, OH), 7.37 (d, IH, J = 5.2 Hz, thiopheneH), 6.74 (d, IH, J= 5.2 Hz, thiopheneH), 3.89 (s, 3H, OMe).
Example 3: O-(2-Methoxycarbonyl-3-thienyl)- NJV-diethylthiocarbamate
Figure imgf000071_0002
[0209] 3-Hydroxy-2-methoxycarbonylthiophene (2.0 g; 12.6 mmol) was dissolved in 10 mL dry DMF and sodium hydride (~60% in mineral oil, 610 mg; ~15.2 mmol) was added portion wise. After hydrogen evolution had ceased the mixture was cooled to 5 °C and diethylthiocarbamoyl chloride (2.48 g; 16.4 mmol) was added in one portion and the temperature was raised to 80 0C for 1 hour. After cooling to room temperature the mixture was poured into a 1% aqueous potassium hydroxide solution (25 mL). The aqueous phase was extracted with toluene (3 times 15 mL). The combined organic phases were washed with 5% aqueous HCl (10 mL), water (10 mL) before drying over Na2SO4. After filtration the solvent was removed by evaporation to give a crude brown oil that was purified by silica gel column chromatography (20 % EtOAc in heptane, R/= 0.45) to give the title compound as a brown oil (1.74 g; 51 %)
[0210] LCMS m/z 274 [M+l]+, tR = 3.98 min, purity (UV/MS): 80/70. GCMS m/z 273, tR = 7.54 min. 1H NMR (CDCl3, 400 MHz) δ 7.44 d, IH, J = 5.6 Hz, thiopheneH), 6.93 (d, IH, J= 5.6 Hz, thiopheneH), 3.88 (q, 2H, J= 7.2 Hz, CH2), 3.82 (s, 3H, OMe), 3.74 (q, 2H, J = 7.2 Hz, CH2), 1.35 (t, 3H, J = 7.2 Hz, Me), 1.33 (t, 3H, J = 7.2 Hz, Me).
Example 4: S-(2-Methoxycarbonyl-3-thienyl)-N,N-diethylcarbamate
Figure imgf000072_0001
[0211] 0-(2-Methoxycarbonyl-3-thienyl)-N,N-diethylcarbamate (1.74 g; 6.4 mmol) was stirred at 22O0C for 30 min. After cooling to room temperature the title compound was obtained as a brown oil. Used without further purification.
[0212] GCMS m/z 273, tR = 7.71 min.
[0213] 1H ΝMR (CDCl3, 400 MHz) δ 7.49 (d, IH, J = 5.2 Hz, thiopheneH4), 7.37 (d, IH, J = 5.2 Hz, thiopheneH5), 3.86 (s, 3H, OMe), 3.55 - 3.39 (m, 4H, 2 x CH2), 1.40 - 1.12 (m, 6H, 2 x CH3).
Example 5: 3-Mercaptothiophene-2-carboxylic acid
Figure imgf000072_0002
[0214] For a published procedure on synthesis of 3-mercaptothiophene-2- carboxylic acid see Corral et al, Synthesis, 1984, 172, which is hereby incorporated by reference in its entirety. 5-(2-Methoxycarbonyl-3-thienyl)-N,N-diethylcarbamate (~6.4 mmol) was refluxed in a mixture of 10% aqueous NaOH (8 mL) and MeOH (4 mL) for 1 hour. After cooling to room temperature MeOH was removed under reduced pressure and the aqueous phase was acidified with 4M HCl. The title compound was isolated by filtration as a light brown solid (670 mg; 65 % over two steps). Used without further purification.
[0215] 1H ΝMR (DMSO-J6, 400 MHz) δ 7.75 (d, IH, J = 5.2 Hz, thiopheneH), 7.07 (d, 2H, J= 5.2 Hz, thiopheneH), 2.48 (s, IH, SH).
Example 6: 3-(4-Ethoxycarboxy-2-nitro-phenylsulfanyl')thiophene-2-carboxylic acid
Figure imgf000073_0001
[0216] Ethyl 4-fluoro-3-nitrobenzoate (440 mg; 2.07 mmol) was dissolved in dry DMF (5 mL) and heated to 700C with cesium carbonate (715 mg; 2.2 mmol). A solution of 3-mercaptothiophene-2-carboxylic acid (300 mg; 1.88 mmol) in dry DMF (5 ml) was added slowly. The mixture was kept at 70 0C for 1 hour before cooling to room temperature overnight. Water was added and the aqueous phase was acidified with 4M HCl before extraction with EtOAc (2 times 10 mL). The combined organic phases were washed thoroughly with water and dried over Na2SO4. After filtration the crude yellow compound was obtained by evaporation of the solvent. Purification was done by silica gel column chromatography (5 % MeOH in DCM, R/= 0.42 in 10 % MeOH in DCM) to give 572 mg (86 %) of a yellow foam.
[0217] LCMS m/z 354 [M+H]+, 371 [M+NH4]+, tR = 7.83 min, purity (UV/MS): 97/30.
[0218] 1H NMR (CD3OD, 400 MHz) δ 8.59 (s, IH, ArH), 7.98 - 7.66 (m, 2H, ArH), 7.20 - 6.95 (m, 2H, ArH), 4.33 (q, 2H, J = 6.8 Hz, OCH2), 1.34 (t, 3H, J= 6.8 Hz, Me). 13C NMR (CD3OD, 100 MHz) δ 165.1, 164.4, 162.8, 146.3, 143.6, 136.3, 135.0, 133.8, 132.8, 130.1, 128.8, 126.9, 62.5, 14.1.
Example 7: 3-(4-Carboxy-2-nitrophenylsulfanyl')thiophene-2-carboxylic acid
Figure imgf000074_0001
[0219] 3-(4-Ethoxycarboxy-2-nitro-phenylsulfanyl)thiophene-2-carboxylic acid (565 mg; 1.6 mmol) was dissolved in 15 mL THF and 10 mL IM aqueous LiOH was added. The solution was heated to 70 0C for 1 hour before cooling to room temperature. TLC (10 % MeOH in DCM) shows no more starting material and only a large orange spot on the baseline. THF was removed by evaporation and after acidification of the aqueous solution using 4M HCl the title compound precipitates as a yellow solid. 435 mg was isolated by filtration (84 %).
[0220] 1H NMR (CD3OD, 400 MHz) δ 8.73 (d, IH, J = 2.0 Hz, ArH), 8.02 (dd, IH, J= 2.0, 8.4 Hz, ArH), 7.85 (d, IH, J= 5.0 Hz, thiopheneH), 7.17 (d, IH, J= 5.0 Hz, thiopheneH), 7.13 (d, IH, J= 8.4 Hz, ArH).
Example 8: 3-(2-Amino-4-carboxyphenylsulfanyl*)thiophene-2-carboxylic acid
Figure imgf000074_0002
[0221] 3-(4-Carboxy-2-nitrophenylsulfanyl)thiophene-2-carboxylic acid (435 mg; 1.34 mmol) was dissolved in 15 mL EtOH. A solution Of K2CO3 (926 mg; 6.7 mmol) and Na2S2O4 (1.37 g; 6.7 mmol) in 10 mL water was added slowly. 5 minutes after ended addition the color changed from orange to light yellow indicating that the nitro compound was reduced to the desired amino compound. After another 15 minutes stirring EtOH was removed by evaporation. The resulting aqueous phase was acidified using 4M HCl before extraction with EtOAc (4 times 20 mL). The combined organic phases were washed with water, dried over Na2SO4, filtered and the solvent removed by evaporation to give 340 mg (86 %) of the title compound. The product was used without further purification.
[0222] 1H NMR (CD3OD, 400 MHz) δ 7.56 (d, IH, J = 2 Hz, ArH), 7.50 - 7.48 (m, 2H, J= 5.6, 8.0 Hz, ArH, thiopheneH), 7.36 (dd, IH, J= 2.0, 8.0 Hz, ArH), 6.33 (d, IH, J= 5.6 Hz, thiopheneH).
Example 9: 10-Oxo-9,10-dihvdro-1.4-dithia-9-azabenz[/1azulene-7-carboxylic acid
Figure imgf000075_0001
[0223] 3-(2-Amino-4-carboxyphenylsulfanyl)thiophene-2-carboxylic acid (340 mg; 1.15 mmol) was dissolved in 5 mL THF. 1,1-Carbonyldiimidazole (630 mg; 3.5 mmol) was added portionwise and stirring was continued at room temperature overnight. Water and 4M HCl was added and the resulting white precipitate isolated by filtration (121 mg; 38 %).
[0224] 1H NMR (DMSO-J6, 400 MHz) δ 10.6 (s, IH, NH), 7.93 (d, IH, J = 5.6 Hz, thiopheneH), 7.81 (d, IH, J = 2.0 Hz, ArH), 7.67 (dd, IH, J = 2.0, 8.0 Hz, ArH), 7.56 (d, IH, J= 8.0 Hz, ArH), 7.11 (d, IH, J= 5.6 Hz, thiopheneH).
Example 10: 10-Chloro-l,4-dithia-9-azabenz[/]azulene-7-carboxylic acid butylamide
Figure imgf000075_0002
[0225] 10-Oxo-9,10-dihydro-l,4-dithia-9-azabenz[/]azulene-7-carboxylic acid (1 15 mg; 0.42 mmol) was dissolved in 5 mL dry toluene. DMF (50 μL) and thionyl chloride (5 mL) was added and the mixture stirred at 70 0C overnight. After cooling the solvent were removed by evaporation. Co-evaporation with three times toluene gave the crude dichloride as a light yellow solid that was used immediately without further purification. [0226] A solution of the crude dichloride in 5 mL dry DCM was added to a solution of n-butylamine (100 μL; 1 mmol) in 5 mL DCM at O0C. The temperature was allowed to rise to room temperature over 2 hours before addition of aqueous NH4Cl (sat, 10 mL). The aqueous phase was extracted with EtOAc (3 times 5 mL) and the combined organic phases dried over Na2SO4 before removal of the solvent in vacuo. The title compound was purified by silica gel column chromatography (10 - 30% EtOAc in heptane, Ry= 0.34 in 30% EtO Ac/heptane) to give 57 mg (41 %) of a yellow solid.
[0227] LCMS m/z 351 [M+H]+, tR = 3.33 min, purity (UV/MS): 99/95.
Example 11: 10-(4-Chlorophenyl)-l,4-dithia-9-azabenz[/1azulene-7-carboxylic acid butylamide
Figure imgf000076_0001
[0228] Bis(triphenylphosphine)palladium(II) chloride (5.7 mg; 0.008 mmol) and 10-chloro-l,4-dithia-9-azabenz[/]azulene-7-carboxylic acid butylamide (27 mg; 0.077 mmol) was mixed in 2 mL THF. 4-Chlorophenylzinc iodide (0.5M in THF, 0.4 mL; 0.2 mmol) was added dropwise and the mixture stirred overnight. The reaction mixture turned green upon reaction overnight. Aqueous NH4Cl (sat) was added and extraction performed with EtOAc. The combined organic phases were dried over Na2SO4, filtered and the solvent removed by evaporation. The crude product was purified by silica gel column chromatography (5-15 % EtOAc in toluene, Ry = 0.67 in 30 % EtOAc in toluene) giving a green solid that was pure on TLC but not according to LCMS. Therefore purification was attempted by preparative LCMS to give 1.5 mg of the desired compound.
[0229] LCMS m/z 427 [M+H]+, 429 [M+H+2]+, tR = 5.50 min, purity (UV/MS): 100/90. 1H NMR (CDCl3, 400 MHz) δ 7.91 - 7.87 (m, 2H, ArH), 7.63 (d, IH, J = 2.0 Hz, ArH), 7.57 (dd, IH, J = 2.0, 8.0 Hz, ArH), 7.54 (d, IH, J = 5.6 Hz, thiopheneH), 7.46 - 7.42 (m, 2H, ArH), 7.40 (d, IH, J = 8.0 Hz, ArH), 7.00 (d, IH, J = 5.6 Hz, thiopheneH), 6.06 (br s, IH, NH), 3.49 - 3.43 (m, 2H, NCH2), 1.64 - 1.54 (m, 2H, CH2), 1.47 - 1.36 (m, 2H, CH2), 0.96 (t, 3H, J= 7.2 Hz, CH3).
Example 12: lO-Phenyl-5-thia-l Al l-triazadibenzo[α. J]cvcloheptene-2.3-dicarboxylic acid bis-butylamide
Figure imgf000077_0001
[0230] 2,2,6,6-Tetramethylpiperidine (675 μL; 4 mmol) was dissolved in 20 mL dry THF and the solution cooled to -3O0C before addition of n-butyl lithium (1.1 M in hexane; 3,64 mL; 4 mmol). The mixture was stirred at -30°C for 1A hour before raising the temperature to 0°C for 1 hour. The mixture was cooled to -78°C and a solution of 10- phenyl-5-thia-l,4,l l-triazadibenzo[α,d]cycloheptene (290 mg; 1 mmol) in VA mL THF was added. Stirring was continued for 10 min. before addition of n-butyl isocyanate (340 μL; 3 mmol). The mixture was stirred at -780C for 2 hours and then quenched by addition of a mixture of 4M HCl (200 μL), water (5 mL) and EtOH (5 mL) and the temperature raised to room temperature. The mixture was neutralized by addition of NaHCO3 (sat) and extraction performed with DCM. The title compound was purified first by silica gel column chromatography (1: 1 EtOAc(heptane, R/= 0.28) followed by recrystallization of the combined fractions from EtOAc to give yellow crystals (25 mg).
[0231] LCMS m/z 488 [M+H]+. 1H NMR (CDCl3, 400 MHz) δ 7.89-7.84 (m, 2H), 7.66-7.44 (m, 5H), 7.41 (dt, IH, J = 1.2, 7.5 Hz), 7.31 (dd, IH, J= 1.6, 7.6 Hz), 7.13 (br t, IH, J= 5.4 Hz), 6.94 (br t, IH, J = 5.4 Hz), 3.51-3.44 (m, 4H), 1.68-1.58 (m, 4H), 1.49-1.37 (m, 4H), 0.95 (dt, 6H, J= 1.2, 7.2 Hz). HPLC tR = 4.84 min.
Example 13: Receptor Selection and Amplification Technology Assay
[0232] The functional receptor assay, Receptor Selection and Amplification Technology (R-SAT®), is used to investigate the pharmacological properties of CBl compounds described herein. R-SAT is disclosed in U.S. Patent Nos. 5,707,798, 5,912,132, and 5,955,281, all of which are hereby incorporated herein by reference in their entirety, including any drawings.
[0233] Briefly, NIH3T3 cells are grown in 96 well tissue culture plates to 70- 80% confluence. Cells are transfected for 16-20 h with plasmid DNAs using Polyfect (Qiagen Inc.) using the manufacturer's protocols. R-SATs are generally performed with 10 ng/well of receptor, 10 ng/well of Gqi5 (Conklin et al, Nature 1993 363:274-6) and 20 ng/well of β-galactosidase plasmid DNA. All receptor constructs are in the pSI-derived mammalian expression vector (Promega Inc). The CBl receptor gene is amplified by PCR from genomic DNA using oligodeoxynucleotide primers based on the published sequence (GenBank Accession # X54937) SEQ ED NO: 1 encodes a CBl receptor truncated after amino acid 417 (SEQ ID NO: 2). The CB2 gene is cloned by performing a PCR reaction on mRNA from spleen. The PCR product containing the entire coding sequence of the CB2 gene is cloned into an expression vector such that the CB2 gene is operably linked to an SV40 promoter. The sequence of the CB2 gene (GenBank Accession #NM_001841) is provided as SEQ ID NO: 3 and the sequence of the encoded CB2 polypeptide is provided as SEQ ID NO: 4. For large-scale transfections, cells are transfected for 16-20 h, then trypsinized and frozen in DMSO. Frozen cells are later thawed, plated at ~10,000 cells per well of a 96 half-area well plate that contained drug. With both methods, cells are then grown in a humidified atmosphere with 5% ambient CO2 for five days. Media is then removed from the plates and marker gene activity is measured by the addition of the β-galactosidase substrate ø-nitrophenyl β-D- galactopyranoside (ONPG) in PBS with 0.5% NP-40. The resulting colorimetric reaction is measured using a spectrophotometric plate reader (Titertek Inc.) at 420 nm. All data is analyzed using the XLFit (IDBSm) computer program. pICso represents the negative logarithm of the concentration of ligand that caused 50% inhibition of the constitutive receptor response. Percent inhibition is calculated as the difference between the absorbance measurements in the absence of added ligand compared with that in the presence of saturating concentrations of ligand normalized to the absorbance difference for the reference ligand (SR141716), which was assigned a value of 100%. These experiments provide a molecular profile, or fingerprint, for each of these agents at the human CB 1 receptor.
[0234] It will be appreciated that the foregoing assay may be used to identify compounds which are agonists, inverse agonists or antagonists of a cannabinoid receptor. In some embodiments, the cannabinoid receptor used in the assay may be a CB 1 receptor. In other embodiments, the cannabinoid receptor used in the assay may consist essentially of SEQ ID NO: 2. In further embodiments, the cannabinoid receptor used in the assay may have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater than at least 99% amino acid identity with a full-length CBl receptor or a truncated CBl receptor of SEQ ID NO: 2.
[0235] Using the following methods, the compounds disclosed herein are evaluated for their ability to bind to a CB 1 receptor. The compounds are tested using a receptor binding assay and then determining of any change in GTPgamma S binding of transfected cells. The results indicate that the compounds are inverse agonists at the CBl receptor.
Example 14: CBl Receptor Binding Assays
[0236] To show that CBl antagonists can block binding of selective CBl ligands to native CBl receptors the ability of compounds of Formula I to block binding of the highly CBl-selective ligand SR1411716 will be examined in rat brain membrane preparations as follows.
[0237] Membrane preparations - Whole brains are harvested from Harlan Sprague Dawley rats and placed in 50 ml Falcon Tubes on ice. The volume is made up to 30 ml with ice-cold membrane buffer (20 mM HEPES, 6 mM MgCl2, 1 mM EDTA, pH 7.2). The Brains are homogenized with a Brinkmann Polytron PT3000 at 20,000 rpm for 40 s. The homogenate is spun at 1,000 x g for 10 min at 40C to remove nuclei and cellular debris. The supernatant is collected and re-centrifuged as previously before membranes are precipitated at 45,000 x g for 20 min at 4°C, resuspended in membrane buffer to a final concentration of 1 mg/ml, snap frozen as aliquots in liquid nitrogen and stored at - 80°C.
[0238] Membrane Binding - 10 μg of membranes are incubated in binding buffer (Ix DMEM with 0.1%BSA) in the presence of 3 nM radioligand ([3H]SR141716A, Amersham Biosciences, Piscataway, NJ) and varying concentrations of ligands (total volume 100 μl in a 96 well plate). Cells are filtered onto a 96 well GF/B filterplate (Packard Bioscience, Shelton, CT) and washed with 300 ml wash buffer (25mM HEPES, 1 mM CaCl2, 5 mM MgCl2, 0.25M NaCl) using a Filtermate 196 Harvester (Packard Instruments, Downers Grove, IL). The filter plates are dried under a heat lamp before addition of 50 μl of scintillation fluid to each well (Microscint 20, Packard, Shelton, CT). Plates are counted on a Topcount NXT (Packard, Shelton, CT).
[0239] Data analysis - Graphs are plotted and KD values are determined by nonlinear regression analysis using Prism software (GraphPad version 4.0, San Diego, CA, USA).
[0240] It will be appreciated that the CB 1 receptor binding assay may be used to identify compounds which are agonists, inverse agonists or antagonists of a cannabinoid receptor. In some embodiments, the cannabinoid receptor used in the assay may be a CBl receptor. In other embodiments, the cannabinoid receptor used in the assay may consist essentially of SEQ ID NO: 2. In further embodiments, the cannabinoid receptor used in the assay may have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater than at least 99% amino acid identity with a full-length CBl receptor or a truncated CBl receptor of SEQ ID NO: 2.
[0241] 10-(4-Chlorophenyl)-l,4-dithia-9-azabenz[/]azulene-7-carboxylic acid butylamide has a pKi 9.0.
Example 15: Sequences for truncated CBl receptors ,
[0242] Below are sequences encoding a truncated CBl receptor. SEO ID NO: !:
ATGAAGTCGATCCTAGATGGCCTTGCAGATACCACCTTCCGCACCATCACCAC
TGACCTCCTGTACGTGGGCTCAAATGACATTCAGTACGAAGACATCAAAGGT
GACATGGCATCCAAATTAGGGTACTTCCCACAGAAATTCCCTTTAACTTCCTT
TAGGGGAAGTCCCTTCCAAGAGAAGATGACTGCGGGAGACAACCCCCAGCTA
GTCCCAGCAGACCAGGTGAACATTACAGAATTTTACAACAAGTCTCTCTCGTC
CTTCAAGGAGAATGAGGAGAACATCCAGTGTGGGGAGAACTTCATGGACATA
GAGTGTTTCATGGTCCTGAACCCCAGCCAGCAGCTGGCCATTGCAGTCCTGTC
CCTCACGCTGGGCACCTTCACGGTCCTGGAGAACCTCCTGGTGCTGTGCGTCA
TCCTCCACTCCCGCAGCCTCCGCTGCAGGCCTTCCTACCACTTCATCGGCAGC
CTGGCGGTGGCAGACCTCCTGGGGAGTGTCATTTTTGTCTACAGCTTCATTGA
CTTCCACGTGTTCCACCGCAAAGATAGCCGCAACGTGTTTCTGTTCAAACTGG
GTGGGGTCACGGCCTCCTTCACTGCCTCCGTGGGCAGCCTGTTCCTCACAGCC
ATCGACAGGTACATATCCATTCACAGGCCCCTGGCCTATAAGAGGATTGTCA
CCAGGCCCAAGGCCGTGGTGGCGTTTTGCCTGATGTGGACCATAGCCATTGT
GATCGCCGTGCTGCCTCTCCTGGGCTGGAACTGCGAGAAACTGCAATCTGTTT
GCTCAGACATTTTCCCACACATTGATGAAACCTACCTGATGTTCTGGATCGGG
GTCACCAGCGTACTGCTTCTGTTCATCGTGTATGCGTACATGTATATTCTCTG
GAAGGCTCACAGCCACGCCGTCCGCATGATTCAGCGTGGCACCCAGAAGAGC
ATCATCATCCACACGTCTGAGGATGGGAAGGTACAGGTGACCCGGCCAGACC
AAGCCCGCATGGACATTAGGTTAGCCAAGACCCTGGTCCTGATCCTGGTGGT
GTTGATCATCTGCTGGGGCCCTCTGCTTGCAATCATGGTGTATGATGTCTTTG
GGAAGATGAACAAGCTCATTAAGACGGTGTTTGCATTCTGCAGTATGCTCTG
CCTGCTGAACTCCACCGTGAACCCCATCATCTATGCTCTGAGGAGTAAGGAC
CTGCGACACGCTTTCCGGAGCATGTTTCCCTCTTGTGAAGGCTAG
SEO ID NO:2
MKSILDGLADTTFRTITTDLLYVGSNDIQYEDIKGDMASKLGYFPQKFPLTSFRGS PFQEKMTAGDNPQLVPADQVNITEFYNKSLSSFKENEENIQCGENFMDIECFMVL NPSQQLAIA VLSLTLGTFTVLENLLVLCVILHSRSLRCRPSYHFIGSLAVADLLGSVI FVYSFIDFHVFHRKDSRNVFLFKLGGVTASFTASVGSLFLTAIDRYISIHRPLAYKRI VTRPKA WAFCLMWTIAIVIA VLPLLGWNCEKLQSVCSDIFPHIDETYLMFWIGVT SVLLLFΓVYAYMYILWKAHSHAVRMIQRGTQKSΠIHTSEDGKVQVTRPDQARMD IRLAKTLVLILVVLIICWGPLLAIMVYDVFGKMNKLIKTVFAFCSMLCLLNSTVNPI IYALRSKDLRHAFRSMFPSCEG*
Example 16: Acute Feeding Study
[0243] Male, Sprague-Dawley rats (90-120 g) serve as subjects for these studies. Rats are fasted for a period of 16 hrs (water was always available). After the fasting period, test compounds are administered either intraperitoneally (ip) or orally (po). Immediately following compound administration, the rats are returned to their home cage. Following 30 min after compound administration, the rats are removed from their home cages and placed individually into clean cages with a pre-measured amount of food. Food weights are obtained (to the nearest 0.1 g) at various time points. Food consumption is monitored for a period of up to 2 hrs (i.e., 2.5 hr after test compound administration).
[0244] Food intake is measured in fasted rats 1 and 2 hours after being administered either 1, 3, or 10 mg/kg doses of a compound described herein.
Example 17: Tail Flick Study
[0245] Male, NSA mice (15-20 g) serve as subjects for these studies. Baseline nociceptive thresholds are assessed using the warm water tail flick test. Briefly, the distal 1/3 to 1A of the tail is immersed in a 520C water bath and the time (to the nearest 0.1 sec) until the mouse removed its tail (i.e., "flicks") from the water is recorded (i.e., tail flick latency). Mice are then injected ip with either vehicle or with various doses of the CBl agonist CP 55,940 and tail flick latencies are recorded for a period of up to 3 hr. A maximum latency of 10 sec is employed in order to prevent tissue damage. In order to determine if a CBl inverse agonists can block the antinociceptive actions of CP 55,940, mice are pretreated with either vehicle or with a test compound 30 min prior to CP55,940. CP55,940 (1 mg/kg) is administered subcutaneously, and the test compound is administered intraperitoneally. Tail flick latencies are then obtained at various time points for a period of up to 2 hr. The vehicle for both compounds is 1:1: 18 cremphor:ethanol:saline.
Example 18: Hypothermia Study
[0246] Male, NSA mice (15-20 g) serve as subjects for these studies. In order to determine if the test compound can block hypothermia elicited by CP 55,940 (1 mg/kg, ip), mice are pretreated with either vehicle or with test compound 30 min prior to CP55,940. Core body temperatures are then obtained at various time points following CP 55,940 administration. Core body temperature (to the nearest 0.10C) is obtained by rectal probe.
Example 19: Chronic Feeding Study
[0247] Male, obese Zucker rats (400-500 g) serve as subjects for these studies. Rats are housed individually and have access to food and water ad libitum. Rats are allowed to acclimate to the vivarium for a period of 3 days, during which body weight and consumption of food and water is monitored. Rats are weighed daily at 1500 hr and then injected with either vehicle or with various doses of the test compound. Daily food and water intakes are also monitored. Food and water bottles are weighed at the time body weights are recorded (i.e., 1350 hr). Vehicle or compound is administered daily for a period of up to 15 days. The test compound attenuates the food and water intake of the rats. Moreover, the attenuation of the food and water intake is dose-dependent.
Example 20: Novel Object Recognition Study
[0248] Subjects: Subjects are male, C57 BK/6 mice, weighing 15-20g upon arrival. Animals are housed 8 per cage with food and water available ad libidum. Animals are housed on a 12 hr light cycle (lights on 6 am) for 4-7 days prior to behavioral testing.
[0249] Equipment: Novel object recognition (NOR) is conducted in a novel environment consisting of a white plastic tub measuring 45.7 x 33.7 x 19 cm. Prior to each trial the bottom of the tub is covered with a piece of plastic lined bench top paper. There are two sets of identical objects chosen so that when given a opportunity to explore, mice would evenly divide exploration time between the objects. "A" objects are yellow, ceramic, 12-sided ramekins measuring 4 cm high x 7 cm diameter. "B" objects are 8 X 8 x 4 cm stainless steel, 4-sided ramekins.
[0250] Procedure: At the beginning of each test day, animals are placed in groups of 6 into clean cages. Testing is conducted in three phases: acclimation, sample and test. For acclimation, each group of six mice is placed collectively into the NOR chamber and allowed to explore freely for 30 min. After acclimation animals are injected (dose and pretreatment time varied by test drug) and placed back into the cages to wait the pre-treatment interval. After the pre-treatment time elapsed, each mouse is placed, one at a time into the NOR chamber, into which two identical objects have been placed ("A" or "B" objects described above). Objects are placed on diagonal corners of the long axis of the arena approximately 5 cm from the walls, while subjects are placed into one of the neutral corners (alternating across subjects). Each mouse is allowed to explore the chamber and the objects for 3 min., and the time spent exploring at each position is recorded. Directly sniffing or touching the object is recorded as exploration. After 3 min., each mouse is removed from the arena and placed back into its cage. The test phase was conducted 1 or 2 hours after the sample phase. During test, one familiar object (seen during sample) and one novel object are placed into the chamber in the same positions used during the sample phase, and each mouse is allowed 3 min to explore. The test sessions are recorded on video and scored by an observer blind to each subject's treatment condition. Any time spent directly sniffing or touching an object was counted as exploration. The object serving as the novel object and the position where the novel object is placed are counterbalanced across subjects. Prior to each trial (acclimation, sample and test), all equipment is wiped with a Clorox wipe and bench paper (cut to fit) is placed in the bottom of the chamber. The procedure is shown below in Scheme 13.
[0251] Measures: In addition to time spent exploring each object (TN = time spent exploring novel object, TF = time spent exploring familiar object), two measures are determined for each subject: exploration ratio (% of time spent exploring at novel object) ER = TN* 100/(TN + Tp) and discrimination index (preference for novel) DI = (TN-TF)/(TN + TF). Scheme 13
Group Acclimation - 30 min. (6 mice/ Group)
Trea 1tment
I P re-treatm ent tim e
Sample Phase - 3 min.
Figure imgf000085_0001
Test Phase - 3 min.
Example 21 : Rotation Study
[0252] Subjects: Subjects are male, Sprague-Dawley rats purchased from Harlan Laboratories, weighing 250-275 g upon arrival. Prior to surgery animals are housed two per cage. All subjects have free access to food and water available for the duration of the study. Animals are housed on a 12 hr light cycle (lights on 6 am), and are acclimated to vivarium conditions for a minimum of one week prior to surgery. All experiments are conducted in accordance with NIH Guidelines for the Care and Use of Laboratory Animals and are approved by the Institutional Animal Care and Use Committee at ACADIA Pharmaceuticals, Inc.
[0253] Surgery. One week after arrival, subjects undergo stereotaxic surgery to unilaterally lesion dopamine terminals within the substantia nigra, a common model of Parkinson's disease. In order to protect noradrenergixc terminals, subjects are administered desipramine (20 mg/kg ip) approximately 20 min prior to surgery. Surgery is conducted under ketamine (80 mg/kg ip) and xylazine (12 mg/kg ip) anesthesia. Animals are placed in the stereotaxic instrument with the incisor bar at -3.2 mm and a hole is drilled in the skull over the substantia nigra according to the atlas of Paxinos and Watson (1997): A/P -5.2 mm, M/L - 2.1 mm. A computer-controlled microsyringe is lowered to -8.2 mm from bregma. 8 μg of 6-hydroxy-dopamine in 4 μl of saline with 0.2% ascorbic acid is infused over 5 min, and 1 min is allowed for diffusion before the syringe is removed and the incision closed. Animals are given a minimum of 15 days after surgery before any behavioral assessment.
[0254] Rotational Behavior. All animals are assessed for rotational behavior in rotometers purchased from San Diego Instruments, Inc. For each behavioral session, subjects are placed in the rotometers and allowed thirty minutes for acclimation. After 30 min., subjects are injected with either the dopamine agonist apomorphine (0.05, 0.16 or 0.5 mg/kg ip in saline with 0.2% ascorbic acid) or the cannabinoid 1 receptor inverse agonist Compound II, N-(butyl)-l l-(4-chlorophenyl)-dibenzo[b,f,][l,4]thiazepine-8- carboxamide, (3 mg/kg in sesame oil). When subjects receive combinations of the two treatments, Compound II is injected 30 minutes prior to apomorphine. After treatment, rotations is measured for 60 min. Subjects are then removed from the rotometers and returned to their home cages. All animals receive all three doses of apomorphine, and the combination of Compound II with both 0.05 mg/kg and 0.16 mg/kg apomorphine. A minimum of 2 days separated test days.
[0255] Although the invention has been described with reference to the above examples, it will be understood that modifications and variations are encompassed within the spirit and scope of the invention. Accordingly, the invention is limited only by the following claims.
References:
[0256] The following references are incorporated by reference herein in their entirety:
1. Le Foil B, Goldberg SR. Cannabinoid CBl receptor antagonists as promising new medications for drug dependence. J Pharmacol Exp Ther. 2005 Mar; 312(3):875-83.
2. Boyd ST, Fremming BA. Rimonabant-a selective CBl antagonist. Ann Pharmacother. 2005 Apr; 39(4):684-90. 3. Howlett AC, Breivogel CS, Childers SR, Deadwyler SA, Hampson RE, Porrino LJ. Cannabinoid physiology and pharmacology: 30 years of progress. Neuropharmacology. 2004; 47 Suppl 1:345-58.

Claims

WHAT IS CLAIMED IS:
Figure imgf000088_0001
as a single isomer, a mixture of isomers, a racemic mixture of isomers, or a pharmaceutically acceptable salt, solvate, metabolite, prodrug, or polymorph thereof, wherein:
X is selected from the group consisting of O, S, S=O, SO2, NRh NC≡N, NC(=Z)Ri,
Figure imgf000088_0002
CRIaRIb1 C=O, C=CRiaRib, and SiRiaRib;
Y is -N(R2) — or -C(R, R2) — ; the symbol =^= represents a single or double bond, where when = is a double bond, R2 is absent;
A is selected from the group consisting of halogen, -NRiaRib, and - N=CRi aRib; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, (cycloalkyl)alkyl, (cycloalkenyl)alkyl, (cycloalkynyl)alkyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, sulfonyl, and -(CH2)O-4-C(^Z)-ORi; provided that A cannot be a substituted or unsubstituted piperazine;
Ari and Ar2 are separately selected from the group consisting of aryl and heteroaryl, each optionally substituted with one or more radical selected from the group consisting of halogen, hydroxyl, nitro, -CN, -C(=Z)Ri, -C(=Z)0Ri, -C(=Z)NR,aR,b, -C(=Z)N(Ri)NR,aRlb, -C(=Z)N(R,)N(R,)C(=Z)R1, -C(R,)=NR,, -NR,.R,b, -N=CR,aR,b, -N(Ri)-CC=Z)R1, -N(R,)-C(=Z)NR,aRlb, -S(O)NRlaRlb, - S(O)2NRlaRib, -N(Ri)-SC=O)R1, -N(R,)-S(=0)2Ri, -OR1, -SR1, and -OCC=Z)R1; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, haloalkyl, and haloalkoxy; and mono-substituted, poly- substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, and sulfonyl; provided that at least one of Ari and Ar2 is an optionally substituted heteroaryl;
D is part of Ari and is selected from the group consisting of CRi, NR2, S, and O;
B is attached to Ari on the carbon adjacent to D and separated from Y by three atoms, wherein one of the three atoms is D, and is selected from the group consisting of halogen, hydroxyl, nitro, -CN, -C(=Z)Ri, -C(=Z)ORi, -C(=Z)NR,.R,b, -C(=Z)N(R,)NRlaRlb, -C(=Z)N(R,)N(R,)C(=Z)R,, -C(Ri)=NR1, -NR,,R,b, -N=CR,aRib, -N(R,)-C(=Z)R,, -N(R,)-C(=Z)NRlaR,b, -S(O)NRlaRlb, - S(O)2NRlaRIb, -N(R,)-S(=O)Ri, -N(R,)-S(=O)2Ri, -OR1, -SR1, and -OC(=Z)R,; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, haloalkyl, and haloalkoxy; and mono-substituted, poly- substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, and sulfonyl; provided that B is not selected from the group consisting of -CF3, phenyl,
-OS(O)2-CF3, methyl, -CN, halogen, and
Figure imgf000089_0001
when A is a substituted or unsubstituted heteroalicyclyl containing at least one nitrogen, cycloalkyl, cycloalkenyl, phenyl, heteroaryl, or -NRiaRib; further provided that B is not halogen when A is substituted or unsubstituted aryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroaryl, halogen, or substituted or unsubstituted sulfenyl; X is - NH; and Y is -N=; Z is O or S;
Ri1 Ria and R^ are each independently selected from the group consisting of hydrogen, halogen, -C(=Z)R3, -C(=Z)OR3, and -C(=Z)NR3aR3b; mono- substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, and haloalkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, -(CH2)0-7-OR3, -(CH2)O-7-SR3, and - (CH2)0-7-NR3aR3b; or Ri a and R^ can be taken together to form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms or an unsubstituted or substituted carbocyclyl having 3 to 9 carbon atoms;
R2 is absent or is selected from the group consisting of hydrogen; mono- substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, and alkynyl; and mono-substituted, poly- substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl;
R3, R3a, and R3b are each independently selected from the group consisting of: hydrogen; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, aryl, heteroaryl, and heteroalicyclyl; and further providing that: when X is O or -NCH3 and Y is -N=, then B cannot be -C(=Z)OH, - C(=Z)Me or-C(=Z)Et; when X is CRuRib and A is phenyl, then B cannot be NH2; when both Ar, and Ar2 are pyridinyl rings, then X cannot be NRi in which Ri is selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, trihalomethyl and hydroxyalkyl; when X is S or NRi and Ri is hydrogen or alkyl, then A cannot be a phenyl ring substituted at the para-position with -CO2H or CO2(alkyl); when X is CRiaRib or C=CRi3Rn,, then Ru and R^ cannot be a heteroalicyclyl or piperazine ring, and Ri a and R^ cannot be taken together to a form a heteroalicyclyl or piperazine ring; when X is NRi and Ri is hydrogen, -C(=O)H or -C(=O)CF3, then A cannot be a phenyl ring substituted at the para-position with a nitro group; when X is NRi and A is aryl, then B cannot be alkyl, alkoxy, hydroxy, or an acid salt thereof; and when X is NRi and Y is -N(R2) ^^, wherein ^= represents a single bond, then B cannot be alkyl or alkoxy.
2. The compound of Claim 1, wherein the compound of Formula (I) binds to a cannabinoid receptor.
3. The compound of Claim 2, wherein the cannabinoid receptor is a CBl receptor.
4. The compound of any one of Claims 1 to 3, wherein Ria and R^ form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms that is optionally substituted with subtituents selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S- sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfϊnyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino.
5. The compound of any one of Claims 1 to 4, wherein Ri3 and Rn, form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from the group consisting of:
Figure imgf000092_0001
wherein R4 and R5 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxyl, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfϊnyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino.
6. The compound of any one of Claims 1 to 5, wherein X is S.
7. The compound of any one of Claims 1 to 6, wherein B is selected from the group consisting of halogen, -CC=Z)R1, -C(=Z)ORi, -C(=Z)NRiaR)b, -C(=Z)N(R,)NRlaRlb, -C(=Z)N(Ri)N(R,)C(=Z)R,, -C(RO=NR1, -NR,,R,b, -N=CRlaRlb, -N(R,)-C(=Z)R,, -N(R,)-C(=Z)NRlaRlb, -S(O)NRlaRlb, -S(O)2NRlaRlb, -N(R,)-S(=O)R,, -N(Ri)-S(=O)2Ri, and -OC(=Z)Ri; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: heteroaralkyl and (heteroalicycyl)alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: aryl, heteroaryl, aralkyl, and heteroalicyclyl.
8. The compound of any one of Claims 1 to 6, wherein B is selected from the group consisting of hydroxyl, -C(=Z)ORi, -C(=Z)N(Ri)NRiaRib, -C(=Z)N(R,)N(R,)C(=Z)R1, -C(RO=NR1, -NR,,Rlb, -N=CRlaRlb, -S(O)NR,,Rlb, -N(RO-SC=O)R1, -N(Ri)-SC=O)2Ri, and -OC(=Z)Ri; mono-substituted, poly-substituted, or unsubstituted, straight or branched chain variants of the following residues: aralkyl, heteroaralkyl, (heteroalicycyl)alkyl, and haloalkoxy; and mono-substituted, poly¬
pi- substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfϊnyl, and sulfonyl.
9. The compound of any one of Claims 1 to 6, wherein B is -C(=Z)NRiaRib
10. The compound of any one of Claims 1 to 6, wherein B is -C(=Z)Ri.
11. The compound of any one of Claims 1 to 6, wherein B is -S(O)2NRiaRib-
12. The compound of Claim 1, wherein if B is -S(O)NRi8Rn,, -S(O)2NR,aRib, -C(=Z)NRiaRib or -C(=Z)N(Ri)NRuRib then Ri, Ria and Rib are each independently selected from the group consisting of:
Figure imgf000093_0001
wherein: n is an integer selected from the group consisting of O, 1, 2, 3, 4, 5, 6 or 7 defining the number of optionally substituted carbon atoms;
Q is selected from the group consisting Of-N(R4)-, O and S;
R4 and R5 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino; and
R6, R6a, Rόb, Rόc, and R6(I are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino; or wherein the substituents selected from the group consisting of R6, R63, R$b, R6O and R^a can be taken together to form a cycloalkyl, cycloalkenyl, cycloalkynyl, or heteroalicyclyl ring with one or more adjacent members of said group consisting Of R6, R68, R<$b, R6C, and R6d.
13. The compound of Claim 12, wherein B is -C(=Z)NRiaRib.
14. The compound of Claim 12, wherein B is -C(=Z)Ri.
15. The compound of Claim 12, wherein B is -S(O)2NRi3RIb.
16. The compound of any of Claims 12 to 15, wherein n is 0, 1, or 2.
17. The compound of any one of Claims 1 to 16, wherein each Ri1 Ri3, Rn,, R2, R3, R3a, R3b are independently selected from aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl, and are substituted with zero to five substituents, wherein each substituent is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino.
18. The compound of any one of Claims 1 to 16, wherein Ri1 Ri3 and Rib are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl.
19. The compound of any one of Claims 1 to 16, wherein Ri and Ru are independently selected from the group consisting of alkyl, cycloalkyl, heteroaryl, heteroalicyclyl and heteroaralkyl; and Rn, is hydrogen.
20. The compound of any one of Claims 1 to 19, wherein A is selected from the group consisting of halogen, -NRiaRib, and -N=CRiaRib; mono-substituted, poly- substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl, alkynyl, (cycloalkyl)alkyl, (cycloalkenyl)alkyl, (cycloalkynyl)alkyl, aralkyl, heteroaralkyl, and (heteroalicycyl)alkyl; and mono- substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, sulfenyl, sulfinyl, sulfonyl, and -(CH2)0-4-C(=Z)-ORi.
21. The compound of any one of Claims 1 to 19, wherein A is an aryl, heteroaryl, or heteroalicyclyl, and is substituted with zero to five substituents, wherein each substituent is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloa'lkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S- sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino, and protected amino.
22. The compound of any one of Claims 1 to 19, wherein A is an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent is independently selected from the group consisting of alkyl, alkoxy, ester, cyano, and halogen.
23. The compound of any one of Claims 1 to 19, wherein A is alkyl or aryl.
24. The compound of any one of Claims 1 to 19, wherein A is cycloalkyl.
25. The compound of Claim 1, wherein: X is S; and
A is selected from the group consisting of a mono-substituted, poly- substituted, or unsubstituted, straight or branched chain variants of the following residues: alkyl, alkenyl and alkynyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl and heteroaralkyl.
26. The compound of Claim 25, wherein A is selected from the group consisting of a mono-substituted, poly-substituted, or unsubstituted, straight or branched alkyl; and mono-substituted, poly-substituted or unsubstituted variants of the following residues: cycloalkyl, aryl, heteroaryl and heteroalicyclyl.
27. The compound of Claim 25, wherein A is a mono-substituted, poly- substituted, or unsubstituted, straight or branched alkyl; or mono-substituted, poly- substituted or unsubstituted aryl.
28. The compound of any one of Claims 25 to 27, wherein B is -C(=Z)NRlaRlb.
29. The compound of any one of Claims 25 to 27, wherein B is -C(=Z)Ri.
30. The compound of one of Claims 28 to 29, wherein Z is O.
31. The compound of any one of Claims 25 to 27, wherein B is - S(O)2NRlaRlb.
32. The compound of any one of Claims 25 to 31, wherein Y is -N(R2) =^; = represents a double bond; and R2 is absent.
33. The compound of any one of Claims 25 to 32, wherein Ri1 Ru and R^ are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl and (heteroalicyclyl)alkyl.
34. The compound of any one of Claims 25 to 32, wherein Ri and Ru are independently selected from the group consisting of alkyl, cycloalkyl, heteroalicyclyl and heteroaralkyl; and R^ is hydrogen.
35. The compound of any one of Claims 1 to 34, wherein Ari is phenyl.
36. The compound of any one of Claims 1 to 34, wherein Ari is selected from the group consisting of optionally substituted pyrazine, optionally substituted pyridine, optionally substituted pyrimidine, optionally substituted oxazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted pyrrole, optionally substituted imidazole, and optionally substituted phenyl.
37. The compound of any one of Claims 1 to 36, wherein Ar2 is selected from the group consisting of optionally substituted pyrazine, optionally substituted pyridine, optionally substituted indole, optionally substituted pyrimidine, optionally substituted oxazole, optionally substituted thiazole, optionally substituted furan, optionally substituted thiophene, optionally substituted pyrrole, optionally substituted imidazole, optionally substituted triazole, optionally substituted isoxazole, optionally substituted isothiazole, optionally substituted pyrazole, and optionally substituted phenyl.
38. The compound of any one of Claims 1 to 37, further comprising a detectable label.
39. The compound of Claim 38, wherein the detectable label is a radiolabel.
40. The compound of Claim 39, wherein the radiolabel is selected from the group consisting of [3H], [18F], [11C] and [125I].
41. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
40 41 42
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
42. A pharmaceutical composition, comprising a therapeutically effective amount of at least one compound of any one of Claims 1 to 41, and a pharmaceutically acceptable carrier, diluent, or excipient.
43. A method of ameliorating or treating a disease or condition selected from the group consisting of obesity, metabolic syndrome, a metabolic disorder, hypertension, polycystic ovary disease, osteoarthritis, a dermatological disorder, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis, a sleep disorder, hyperlipidemic conditions, bulimia nervosa, a compulsive eating disorder, an appetite disorder, atherosclerosis, diabetes, high cholesterol, hyperlipidemia, cachexia, an inflammatory disease, rheumatoid arthritis, a neurological disorder, a psychiatric disorder, substance abuse, depression, anxiety, mania, schizophrenia, dementia, dystonia, muscle spasticity, tremor, psychosis, an attention deficit disorder, a memory disorder, a cognitive disorder, short term memory loss, memory impairment, drug addiction, alcohol addiction, nicotine addiction, infertility, hemorrhagic shock, septic shock, cirrhosis, a cardiovascular disorder, cardiac dysfunction, valvular disease, myocardial infarction, cardiac hypertrophy, congestive heart failure, transplant rejection, an intestinal disorder, a neurodegenerative disease, multiple sclerosis, Alzheimer's disease, Parkinson's disease, epilepsy, Huntington's disease, Tourette's syndrome, cerebral ischaemia, cerebral apoplexy, craniocerebral trauma, stroke, spinal cord injury, catabolism, hypotension, hemorrhagic hypotension, endotoxin- induced hypotension, an eye disorder, glaucoma, uveitis, retinopathy, dry eye, macular degeneration, emesis, nausea, a gastric ulcer, diarrhea, pain, a neuropathic pain disorder, viral encephalitis, plaque sclerosis, cancer, a bone disorder, bone density loss, osteoporosis, ostepenia, a lung disorder, asthma, pleurisy, polycystic ovary disease, premature abortion; inflammatory bowel disease, lupus, graft vs. host disease, T-cell mediated hypersensitivity disease, Hashimoto's thyroiditis, Guillain-Barre syndrome, contact dermatitis, allergic rhinitis, ischemic injury, and reperfusion injury comprising administering to a subject a therapeutically effective amount of at least one compound of any one of Claims 1 to 41.
44. The method of Claim 43, wherein the therapeutically effective amount of a compound of Claim 1 is in a sufficient amount to ameliorate or treat said disease or condition by binding to a cannabinoid receptor.
45. The method of Claim 43, further comprising identifying a subject in need of ameliorating or preventing said disease or condition.
46. The method of Claim 43, wherein the neurological disorder is selected from the group consisting of schizophrenia, dementia, dystonia, muscle spasticity, tremor, psychosis, anxiety, depression, an attention deficit disorder, a memory disorder, a cognitive disorder, drug addiction, alcohol addiction, nicotine addiction, a neurodegenerative disease, multiple sclerosis, Alzheimer's disease, Parkinson's disease, epilepsy, Huntington's disease, Tourette's syndrome, cerebral ischaemia, cerebral apoplexy, craniocerebral trauma, stroke, spinal cord injury, pain, neuropathic pain disorder, viral encephalitis, and plaque sclerosis.
47. The method of Claim 43, wherein the disease or condition is selected from the group consisting of obesity, metabolic syndrome, an appetite disorder, cachexia, high cholesterol, hyperlipidemia, and diabetes.
48. The method of Claim 43, wherein the disease or condition is selected from the group consisting of emesis, nausea, a gastric ulcer, diarrhea, and intestinal disorders.
49. The method of Claim 43, wherein said inflammatory disease is selected from the group consisting of rheumatoid arthritis, asthma, and psoriasis.
50. The method of Claim 43, wherein the disease or condition is selected from the group consisting of hemorrhagic shock, septic shock, cirrhosis, atherosclerosis, and a cardiovascular disorder.
51. The method of Claim 43, wherein the disease or condition is selected from the group consisting of infertility and premature abortion.
52. The method of Claim 43, wherein the disease or condition is selected from the group consisting of glaucoma, uveitis, retinopathy, dry eye, and macular degeneration.
53. The method of Claim 43, wherein the disease or condition is selected from the group consisting of osteoporosis and ostepenia.
54. The method of Claim 43, wherein the disease or condition is selected from the group consisting of asthma and pleurisy.
55. The method of Claim 43, wherein the disease or condition is cancer.
56. A method for ameliorating or treating a disease or condition in which it would be beneficial to modulate the activity of a CBl receptor comprising administering to a subject a therapeutically effective amount of a compound of any one of Claims 1 to 41.
57. A method of ameliorating or treating drug addition or alcohol addiction comprising administering to a subject a therapeutically effective amount of a compound of any one of Claims 1 to 41.
58. The method of Claim 57, further comprising identifying a subject in need of ameliorating a drug addiction or alcohol addiction.
59. A method of ameliorating or treating obesity comprising administering to a subject a therapeutically effective amount of a compound of any one of Claims 1 to 41.
60. The method of Claim 59, further comprising identifying a subject in need of ameliorating obesity.
61. A method of ameliorating or treating impaired cognition or a memory disorder comprising administering to a subject a therapeutically effective amount of a compound of Claim 1.
62. The method of Claim 61, further comprising identifying a subject in need of ameliorating impaired cognition.
63. A method of improving cognition or memory in a subject comprising administering to the subject a therapeutically effective amount of a compound of any one of Claims 1 to 41.
64. A method of alleviating or treating inflammation due to an inflammatory disease comprising administering to a subject a therapeutically effective amount of a compound of any one of Claims 1 to 41.
65. The method of Claim 64, wherein the inflammatory disease is selected from the group consisting of rheumatoid arthritis, asthma, and psoriasis.
66. The method of Claim 64, further comprising identifying a subject in need of ameliorating an inflammatory disease.
67. A method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor in a subject comprising administering to the subject an effective amount of a compound of any one of Claims 1 to 41.
68. The method of Claim 67, wherein the cannabinoid receptor is a CBl receptor.
69. A method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor comprising contacting a cannabinoid receptor with a compound of any one of Claims 1 to 41.
70. The method of Claim 69, wherein the cannabinoid receptor is a CBl receptor.
71. A method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor comprising: contacting a cannabinoid receptor with at least one test compound of any one of Claims 1 to 41; and determining any increase or decrease in activity level of the cannabinoid receptor so as to identify said test compound as an agonist, inverse agonist or antagonist of the cannabinoid receptor.
72. The method of Claim 71 , wherein the cannabinoid receptor is a CB 1 receptor.
73. The method of Claim 71, wherein said cannabinoid receptor consists essentially of SEQ ID NO: 2.
74. The method of Claim 71, wherein said cannabinoid receptor has at least 90% amino acid identity to SEQ ID NO: 2.
75. The method of Claim 71, wherein said cannabinoid receptor has at least 85% amino acid identity to SEQ ID NO: 2.
76. The method of Claim 71, wherein said cannabinoid receptor has at least 70% amino acid identity to SEQ ID NO: 2.
77. A method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor comprising: culturing cells that express said cannabinoid receptor; incubating the cells or a component extracted from the cells with at least one test compound of any one of Claims 1 to 41; and determining any increase or decrease in activity of the cannabinoid receptor so as to identify said test compound as an agonist, inverse agonist, or antagonist of the cannabinoid receptor. •78. The method of Claim 77, wherein the cannabinoid receptor is a CBl receptor.
79. The method of Claim 77, wherein said cannabinoid receptor consists essentially of SEQ ID NO: 2.
80. The method of Claim 77, wherein said cannabinoid receptor has at least 90% amino acid identity to SEQ ID NO: 2.
81. The method of Claim 77, wherein said cannabinoid receptor has at least 85% amino acid identity to SEQ ID NO: 2.
82. The method of Claim 77, wherein said cannabinoid receptor has at least 70% amino acid identity to SEQ ID NO: 2.
83. A method for identifying a compound which binds to a CB-I receptor comprising: labeling a compound selected from any one of Claims 1 to 41; with a detectable label; contacting the CB-I receptor with the labeled compound; and determining whether the labeled compound binds to the CB-I receptor.
84. The method of Claim 83, wherein the detectable label is a radiolabel.
85. The method of Claim 84, wherein the radiolabel is selected from the group consisting of [3H], [18F], [1 1C] and [125I].
PCT/US2007/022183 2006-10-17 2007-10-17 Cb1-modulating compounds and their use WO2008048648A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US85257906P 2006-10-17 2006-10-17
US60/852,579 2006-10-17

Publications (2)

Publication Number Publication Date
WO2008048648A2 true WO2008048648A2 (en) 2008-04-24
WO2008048648A3 WO2008048648A3 (en) 2008-06-12

Family

ID=39204737

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/022183 WO2008048648A2 (en) 2006-10-17 2007-10-17 Cb1-modulating compounds and their use

Country Status (1)

Country Link
WO (1) WO2008048648A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010018686A1 (en) * 2008-08-12 2010-02-18 日本曹達株式会社 Nitrogenated heterocyclic derivative, and agricultural or horticultural bactericidal agent
CN102807574A (en) * 2011-05-30 2012-12-05 长春吉大天元化学技术股份有限公司 Novel pyrido sulfur nitrogen 7-membered ring derivatives adopted as anti-tumor drugs, preparation method and applications thereof
US9062024B2 (en) 2009-09-28 2015-06-23 Nippon Soda Co., Ltd. Nitrogen-containing heterocyclic compound and salt thereof, and a fungicide for agricultural and horticultural use
US10112915B2 (en) 2015-02-02 2018-10-30 Forma Therapeutics, Inc. 3-aryl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10183934B2 (en) 2015-02-02 2019-01-22 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10308712B2 (en) 2014-03-27 2019-06-04 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US10555935B2 (en) 2016-06-17 2020-02-11 Forma Therapeutics, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
US11421026B2 (en) 2015-09-30 2022-08-23 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005044822A1 (en) * 2003-11-07 2005-05-19 Pfizer Products Inc. Bicyclic pyrazolyl and imidazolyl compounds and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005044822A1 (en) * 2003-11-07 2005-05-19 Pfizer Products Inc. Bicyclic pyrazolyl and imidazolyl compounds and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HOWLETT A C ET AL: "Cannabinoid physiology and pharmacology: 30 years of progress" NEUROPHARMACOLOGY, PERGAMON PRESS, OXFORD, GB, vol. 47, 2004, pages 345-358, XP004612197 ISSN: 0028-3908 cited in the application *

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010018686A1 (en) * 2008-08-12 2010-02-18 日本曹達株式会社 Nitrogenated heterocyclic derivative, and agricultural or horticultural bactericidal agent
JP5303561B2 (en) * 2008-08-12 2013-10-02 日本曹達株式会社 Nitrogen-containing heterocyclic derivatives and agricultural and horticultural fungicides
KR101315628B1 (en) * 2008-08-12 2013-10-08 닛뽕소다 가부시키가이샤 Nitrogen-containing heterocyclic derivative and fungicide for agricultural and horticultural use
US8557738B2 (en) 2008-08-12 2013-10-15 Nippon Soda Co., Ltd. Nitrogen-containing heterocyclic derivative and fungicide for agricultural and horticultural use
US9062024B2 (en) 2009-09-28 2015-06-23 Nippon Soda Co., Ltd. Nitrogen-containing heterocyclic compound and salt thereof, and a fungicide for agricultural and horticultural use
CN102807574A (en) * 2011-05-30 2012-12-05 长春吉大天元化学技术股份有限公司 Novel pyrido sulfur nitrogen 7-membered ring derivatives adopted as anti-tumor drugs, preparation method and applications thereof
WO2012163236A1 (en) * 2011-05-30 2012-12-06 长春吉大天元化学技术股份有限公司 Pyridothiazepine derivative, preparation method and uses therefor
US10308712B2 (en) 2014-03-27 2019-06-04 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US11566069B2 (en) 2014-03-27 2023-01-31 Bird Rock Bio, Inc. Treatment of disease responsive to modulation of cannabanoid 1(CB1) receptor signaling
RU2730674C2 (en) * 2014-03-27 2020-08-24 Берд Рок Байо, Инк., Antibodies binding to human cannabinoid receptor 1 (cb1)
US10464910B2 (en) 2015-02-02 2019-11-05 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494351B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10214500B2 (en) 2015-02-02 2019-02-26 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10377726B2 (en) 2015-02-02 2019-08-13 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10407418B2 (en) 2015-02-02 2019-09-10 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10414738B2 (en) 2015-02-02 2019-09-17 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10421731B2 (en) 2015-02-02 2019-09-24 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10421732B2 (en) 2015-02-02 2019-09-24 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10428031B2 (en) 2015-02-02 2019-10-01 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10442776B2 (en) 2015-02-02 2019-10-15 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10450284B2 (en) 2015-02-02 2019-10-22 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10450283B2 (en) 2015-02-02 2019-10-22 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10457652B2 (en) 2015-02-02 2019-10-29 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10214501B2 (en) 2015-02-02 2019-02-26 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10464909B2 (en) 2015-02-02 2019-11-05 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10472337B2 (en) 2015-02-02 2019-11-12 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10479772B2 (en) 2015-02-02 2019-11-19 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494352B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494353B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10239845B2 (en) 2015-02-02 2019-03-26 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494354B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10501424B2 (en) 2015-02-02 2019-12-10 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10513501B2 (en) 2015-02-02 2019-12-24 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11891365B2 (en) 2015-02-02 2024-02-06 Valo Health, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10183934B2 (en) 2015-02-02 2019-01-22 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10822316B2 (en) 2015-02-02 2020-11-03 Valo Early Discovery, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10829461B2 (en) 2015-02-02 2020-11-10 Valo Early Discovery, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10829462B2 (en) 2015-02-02 2020-11-10 Valo Early Discovery, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10870645B2 (en) 2015-02-02 2020-12-22 Valo Early Discovery, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US11702412B2 (en) 2015-02-02 2023-07-18 Valo Health, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10988450B2 (en) 2015-02-02 2021-04-27 Valo Early Discovery, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11274085B2 (en) 2015-02-02 2022-03-15 Valo Health, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11274084B2 (en) 2015-02-02 2022-03-15 Valo Health, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11279681B2 (en) 2015-02-02 2022-03-22 Valo Health, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10112915B2 (en) 2015-02-02 2018-10-30 Forma Therapeutics, Inc. 3-aryl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11421026B2 (en) 2015-09-30 2022-08-23 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
US10874649B2 (en) 2016-06-17 2020-12-29 Valo Early Discovery, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
US11730721B2 (en) 2016-06-17 2023-08-22 Valo Health, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
US10555935B2 (en) 2016-06-17 2020-02-11 Forma Therapeutics, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors

Also Published As

Publication number Publication date
WO2008048648A3 (en) 2008-06-12

Similar Documents

Publication Publication Date Title
US20070105819A1 (en) CB-1 modulating compounds and their use
WO2008048648A2 (en) Cb1-modulating compounds and their use
US7514441B2 (en) Substituted pyrazolo [1,5-A]pyrimidines as calcium receptor modulating agents
WO2008118141A2 (en) Use of cannabinoid modulating compounds in combination with other therapeutic compounds for adjunctive therapy
CA2619566C (en) 6-substituted-2,3,4,5-tetrahydro-1h-benzo[d]azepines as 5-ht2c receptor agonists
CS9101168A2 (en) Pharmaceutical mixtures and method of their active substance production
SK96494A3 (en) Piperazine and piperidine derivatives, and their use as antipsychotic
KR101738866B1 (en) Cyclic N,N&#39;-diarylthioureas and N,N&#39;-diarylureas as androgen receptor antagonists, anti-cancer agent, method for producing and using same
WO2004074291A1 (en) Triazole compounds useful in therapy
CA2844525A1 (en) Cyclopropaneamine compound
KR20050085691A (en) 4,5-diarylthiazole derivatives as cb-1 ligands
JP2007522142A (en) Benzimidazole-substituted thiophene derivatives having activity against IKK3
KR20130119854A (en) Pyrazole compounds as sigma receptor inhibitors
SG185484A1 (en) Heteroaryl-cyclohexyl-tetraazabenzo[e]azulenes
JP3156933B2 (en) Fused thiophene compounds and their pharmaceutical uses
US6395735B2 (en) Pharmaceutical compounds
US7271168B2 (en) Piperazine derivatives having SST1 antagonistic activity
CN100374440C (en) Spirosubstituted piperidines as selective melanin concentrating hormone receptor antagonists for the treatment of obesity
EP1725232B1 (en) (indol-3-yl)-heterocycle derivatives as agonists of the cannabinoid cb1 receptor
TW200845973A (en) Heterocycles as orexin antagonists
JP2007533715A (en) Compounds having morpholinyl and piperidinyl groups for use as GlyT1 inhibitors
JP2007505106A (en) Substituted piperazines of azepine, oxazepine, and thiazepine
WO1997003986A1 (en) Fused triazole compounds
US20080090805A1 (en) Cb-1 modulating compounds and their use
KR20100135248A (en) Indolinone compound

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07839649

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07839649

Country of ref document: EP

Kind code of ref document: A2