WO2007047737A1 - Cb-1 modulating compounds and their use - Google Patents

Cb-1 modulating compounds and their use Download PDF

Info

Publication number
WO2007047737A1
WO2007047737A1 PCT/US2006/040662 US2006040662W WO2007047737A1 WO 2007047737 A1 WO2007047737 A1 WO 2007047737A1 US 2006040662 W US2006040662 W US 2006040662W WO 2007047737 A1 WO2007047737 A1 WO 2007047737A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
group
alkyl
heteroalicyclyl
substituted
Prior art date
Application number
PCT/US2006/040662
Other languages
French (fr)
Inventor
Roger Olsson
Fredrik Ek
Lars Korsgaard Ottesen
Anne Bulow
Ethan Burstein
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.
Priority to EP06826157A priority Critical patent/EP1937677A1/en
Priority to CA002625423A priority patent/CA2625423A1/en
Publication of WO2007047737A1 publication Critical patent/WO2007047737A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D419/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms
    • C07D419/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D419/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/14Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D223/18Dibenzazepines; Hydrogenated dibenzazepines
    • C07D223/20Dibenz [b, e] azepines; Hydrogenated dibenz [b, e] azepines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D225/00Heterocyclic compounds containing rings of more than seven members having one nitrogen atom as the only ring hetero atom
    • C07D225/04Heterocyclic compounds containing rings of more than seven members having one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D225/08Heterocyclic compounds containing rings of more than seven members having one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems condensed with two six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/10Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D243/38[b, e]- or [b, f]-condensed with six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D245/00Heterocyclic compounds containing rings of more than seven members having two nitrogen atoms as the only ring hetero atoms
    • C07D245/04Heterocyclic compounds containing rings of more than seven members having two nitrogen atoms as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • C07D245/06Heterocyclic compounds containing rings of more than seven members having two nitrogen atoms as the only ring hetero atoms condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D267/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D267/02Seven-membered rings
    • C07D267/08Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D267/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D267/16Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with two six-membered rings
    • C07D267/20[b, f]-condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D281/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D281/02Seven-membered rings
    • C07D281/04Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D281/08Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D281/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with two six-membered rings
    • C07D281/16[b, f]-condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/192Radicals derived from carboxylic acids from aromatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D337/00Heterocyclic compounds containing rings of more than six members having one sulfur atom as the only ring hetero atom
    • C07D337/02Seven-membered rings
    • C07D337/06Seven-membered rings condensed with carbocyclic rings or ring systems
    • C07D337/10Seven-membered rings condensed with carbocyclic rings or ring systems condensed with two six-membered rings
    • C07D337/14[b,f]-condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D419/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms
    • C07D419/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D419/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

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 CBl.
  • 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 CBl 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). hi 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; Sanofi-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 CBl receptor antagonists and inverse agonists for the treatment of alcohol addiction, opiate addiction, cocaine addiction, anxiety, and septic shock.
  • mice null for the CBl gene also display impaired cocaine 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 CB 1 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.
  • 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 and/or condition that would be alleviated, improved, and/or prevented by administration of a compound that modulates a cannabinoid receptor comprising administering to a therapeutically effective amount of a compound of Formula (I). Also disclosed herein are pharmaceutical compositions comprising a compound of Formula CD-
  • Figure IA is a graph showing the percent response of the CBl receptor as the concentration of l l-Cyclohexyl-dibenzo[b,fj [l,4]thiazepine-8-carboxylic acid piperidin- 1-ylamide (Compound I) increases.
  • Figure IB is a graph showing the percent response of the CB2 receptor as the concentration of Compound I increase.
  • Figure 2 is a bar graph showing the food intake in fasted rats 1 and 2 hours after being administered either 1, 3, or 10 mg/kg doses of Compound I. * Indicates p ⁇ 0.05 as compared to the vehicle-treated controls. ** Indicates p ⁇ 0.01 as compared to the vehicle- treated controls.
  • Figure 3 is bar graph showing the time course food intake in fasted rats after being administered 1 mg/kg of Compound I. * Indicates p ⁇ 0.05 as compared to the vehicle-treated controls. ** Indicates p ⁇ 0.01 as compared to the vehicle-treated controls.
  • Figure 4 is a bar graph showing cumulative food consumption at several points in time after the rats had been dosed with 10 mg/kg of Compound I. * Indicates p ⁇ 0.05 as compared to the vehicle-treated controls.
  • Figure 5 A is a line graph showing the attenuation of CB 1 agonist-mediated effects after administration of CP 55,940 (0.3 and 1.0 mg/kg).
  • Figure 5B is a line graph showing the attenuation of CBl agonist-mediated effects after administration of Compound I alone or in combination with CP55,940.
  • Figure 6 is a bar graph showing the body temperature of the rats at several points in time after the rats had been dosed with various doses of CP 55,950 or CP55,950 and Compound I.
  • Figure 7 is a bar graph showing the concentration of Compound I in the plasma and brain at several points in time.
  • Figures 8 A and 8B are bar graphs showing the concentration of compound, N-(butyl)- 11 -(4-chlorophenyl)-dibenzo [b,f,] [ 1 ,4]thiazepine-8 -carboxamide
  • FIGS 8C and 8D are line graphs showing the concentration of Compound II in the plasma and brain at several points in time.
  • Figure 9B is a line graph showing the effects of Compound II (1 and 3 mg/kg/day) on food intake and water intake.
  • Figure 9C line graph showing the effects of Compound II (10 mg/kg/day) on body weight.
  • Figure 9D is a line graph showing the effects of Compound II (10 mg/kg/day) on food intake and water intake.
  • Figures 1OA and 1OC are bar graphs showing the exploration ratio at 1 and 2 hours after the mice had been dosed with the vehicle, CP 55,940 (0.3 mg/kg, ip), or SR 141716A (I mg/kg, ip).
  • Figures 1OB and 1OD are bar graphs showing the discrimination index at 1 and 2 hours after the mice had been dosed with the vehicle, CP 55,940 (0.3 mg/kg, ip), or SR141716A (1 mg/kg, ip).
  • Figure 1 IA is a bar graph showing the exploration ratio 2 hours after the mice had been dosed with Compound II (3 mg/kg, ip).
  • Figure 1 IB is a bar graph showing the discrimination index 2 hours after the mice had been dosed with Compound II (3 mg/kg, ip).
  • Figure 12 is a bar graph showing percentage of novel recognition of a familiar object 2 hours after the mice had been dosed with 1, 3, or 10 mg/kg of Compound II.
  • Figure 13 is a line graph showing the working memory errors of the mice after being dosed with the vehicle, tacrine (0.3 mg/kg), or Compound II (3 mg/kg).
  • Figure 14 is a line graph showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.05, 0.16, and 0.5 mg/kg).
  • Figure 15 is a line graph showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.05 mg/kg), Compound II (3.0 mg/kg), or apomorphine (0.05 mg/kg) and Compound II (3.0 mg/kg).
  • Figure 16 is a line graph showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.16 mg/kg), Compound II (3.0 mg/kg), or apomorphine (0.16 mg/kg) and Compound II (3.0 mg/kg).
  • Z can be O (oxygen) or S (sulfur);
  • R la and Ri b 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;
  • R 2 can be absent or is selected from the group consisting of: 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;
  • R 3, R 3a , and R 3b can each independently selected from the group consisting of: 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;
  • A cannot be a substituted or unsubstituted piperazine.
  • H cannot be selected from the group consisting of-
  • CF 3 phenyl, -OS(O) 2 -CF 3 , methyl, -CN, halogen, and when A is a substituted or unsubstituted heteroalicyclyl containing at least one nitrogen or-NRi a Ri b .
  • H cannot be halogen when A is substituted or unsubstituted aryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroaryl, halogen, and substituted or unsubstituted sulfenyl;
  • X is -NR 1 , wherein Rj is hydrogen; and
  • the compound of Formula (I) can bind to a cannabinoid receptor.
  • the cannabinoid receptor can be a CBl receptor.
  • Rj 3 and R ⁇ can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms and 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-sul
  • subtituents
  • R 4 and R 5 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, iso
  • Ri 3 and R ⁇ can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from the group consisting of: [0034]
  • Ri b can be hydrogen.
  • R ⁇ can be C 1-3 alkyl.
  • X can be S, SO, or SO 2 .
  • H can be selected from the group consisting of cycloalkyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted.
  • H can be an unsubstituted or substituted heteroaryl is selected from the group consisting of:
  • H can be an optionally substituted phenyl.
  • the optionally substituted phenyl can be substituted with a Ci -4 alkyl.
  • Rj 8 can be selected from the group consisting of alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH 2 ) 0-7 -NR 3a R 3 b, wherein any member of said group can be substituted or unsubstituted.
  • R 1 a can be selected from the group consisting of alkyl, alkoxy, aryl, aralkyl, heteroaryl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted.
  • Ri a can an optionally substituted heteroaryl or heteroaralkyl.
  • the optionally substituted heteroaryl or heteroaralkyl can be hydrogen or methyl.
  • n can be 1 or 2.
  • the optionally substituted heteroaralkyl can be
  • n can be 1 or 2.
  • H can be -N(Rj)-C( ⁇ Z)NRi 3 RIb and Ri is hydrogen and Ri 3 is alkyl or aralkyl.
  • R ⁇ can be hydrogen.
  • H can be -S(O)NRi a Ri b or -S(O) 2 NRi a Rib-
  • H can be -S(O)NRi a Ri b or -S(O) 2 NRi a Ri b and Ri a can be selected from the group consisting of alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl.
  • R ⁇ can be hydrogen.
  • n can be 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 can be selected from the group consisting Of-N(R 4 )-, O and S;
  • R 4 and R 5 each 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, 0-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothi
  • R 6 , R 6a , R ⁇ b, R ⁇ c and Red can 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, iso
  • R 1, Ri a , R 2a , R 2 , R 3 , R 3a> and R 3b can be each independently selected from the group consisting of aryl, heteroaryl, heteroalicyclyl, aralkyl, heteraralkyl, or (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,
  • A can be 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, 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, N-sulf
  • A can be an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent can be independently selected from the group consisting of 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).
  • the phenyl when A is substituted phenyl, the phenyl can be substituted with a halogen, methoxy, or cyano group.
  • A can be selected from the group consisting of 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 unsubstituted;
  • B, C, D, E, F, G and I can be separately selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cyclo
  • Z can be O (oxygen).
  • A can be selected from the group consisting of 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 the group consisting of 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.
  • 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
  • heteroaryl e.g., thiophene
  • C 3 - C 12 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 - NRi a Ri b wherein Rj a is an aryl (e.g.,optionally substituted phenyl) and R ⁇ is hydrogen.
  • A can be -NRi a Rj b wherein Rj 3 is a phenyl group substituted with a halogen and R ⁇ is hydrogen.
  • A can be C 3 -C] 2 alkyl (e.g., n- propyl), C 4 -C 12 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 -, Cg-, Cio-, Cn-, C] 2 alkyl.
  • A is not C 4 -, C 5 -, C 6 -, C 7 -, C 8 -, C 9 -, C 10 -, Cn-, Ci 2 alkyl.
  • A is not cycloalkyl.
  • A is not aryl.
  • A is not heteroaryl.
  • A is not heteroalicyclyl,
  • A is not halogen, -NRi a Ri t ,.
  • X can be S (sulfur); and
  • Y can be -N(R 2 )- wherein the symbol — represents a double bond and R 2 does not exist.
  • X can be S;
  • X can be S;
  • A can be an aryl or a heteroaryl group
  • X can be S
  • A can be a cycloalkyl, a heteroalicyclyl, or -NRi 3 R-Ib group;
  • Rj 3 can be selected from the group consisting of alkyl, alkoxy, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH 2 )o -7 -NR 3a R 3
  • X can be S;
  • X can be S;
  • A can be selected from the group consisting of aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRi a Ri b group;
  • X can be S;
  • X can be S;
  • X can be S;
  • A can be selected from the group consisting of aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRi a Rib group;
  • X can be S;
  • the optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl is selected from the group consisting of:
  • n can be 1 or 2.
  • X can be S;
  • X can be S;
  • A can be selected from the group consisting of aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRi a Ri b group;
  • X can be S;
  • the optionally substituted aryl or aralkyl can be selected from the group
  • n 1 or 2.
  • X can be S;
  • X can be S;
  • A can be selected from the group consisting of aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NR la Rib group;
  • X can be S;
  • heteroalicyclylalkyl can be selected from the group consisting of: ,
  • n can be 1 or 2.
  • X can be S;
  • X can be S;
  • A can be selected from the group consisting of aryl (e.g., unsubstituted or substituted phenyl) or a heteroaryl (e.g., thiophene and pyridine);
  • X can be S;
  • A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NR) a Ri b group;
  • X can be S;
  • the optionally substituted heteroaralkyl is from the
  • n can be 1 or 2.
  • heteroaralkyl wherein Q can be oxygen or sulfur, and in some of the embodiments, n can be 1 or 2.
  • the compound of Formula (I) can be selected from the group consisting of:
  • Certain of the compounds of the present invention may exist as stereoisomers including optical isomers.
  • the invention includes all stereoisomers and both the racerhic 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.
  • Still another embodiment described herein relates to a pharmaceutical composition, comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier, diluent, or excipient.
  • any "R" group(s) such as, without limitation, R], Ri 8 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 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.
  • R 3 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.
  • a substituent is deemed to be “optionally subsituted,” or “substituted” it is meant that the subsitutent 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-sulf
  • 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 "Ci-C 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 the group consisting of 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, 0-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.
  • 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.
  • aryl refers to a carbocyclic (all carbon) ring or two or more fused rings (rings that share two adjacent carbon atoms) that have a fully delocalized pi- electron system.
  • 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, isocyan
  • heteroaryl refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system), 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 aryloxyl 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 Cio, 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 connetected 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 the group consisting of 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 quaternized; 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 the group consisting of 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, thiocyan
  • heteroalicyclic or “heteroalicyclyl” include but ate not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, morpholinyl, oxiranyl, piperidinyl iV-Oxide, piperidinyl, piperazinyl, pyrrolidinyl, 4-piperidonyl, pyrazolidinyl, 2-oxopyrrolidinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, and thiamorpholinyl sulfone.
  • 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, trifluoromethyl 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.
  • 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.
  • 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.
  • 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.
  • 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 “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 defined with respect to O-carboxy.
  • An S-sulfonamido may be substituted or unsubstituted.
  • N-sulfonamido refers to a "RSO 2 N(R A )-" group in which R and RA can be the same as 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.
  • 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).
  • substituents are not specified (e.g. haloalkyl), there may be one or more substituents present.
  • 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 alkoxy groups 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)methyl
  • 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 CB 1 or of a CB 1 subtype 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 was 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 by a medical professional such as an M.D. or a D.V.M. 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.
  • 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.
  • R 1a and R 1b are as defined above for Formula I.
  • R 3 and R 4 can be selected from the same group of substituents as R 1a and R 1b as defined above for Formula I.
  • R la , Rib, and A are as defined above for Formula I.
  • R 3 and R 4 can be selected from the same group of substituents as R 1 ⁇ and Ri b as defined above for Formula I.
  • Ri a , Rib, and A are as defined above for Formula I.
  • R 3 and R 4 can be selected from the same group of substituents as Rj 3 and R ⁇ as defined above for Formula I.
  • Scheme 5
  • Ri a , Rib, and A are as defined above for Formula I.
  • Ri a , Rib, and A are as defined above for Formula I.
  • R 3 and R 4 can be selected from the same group of substituents as Rj a and R ⁇ as defined above for Formula I.
  • 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 preventing 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 the group consisting of: a method of treating or preventing 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, cocaine, hallucinogens, inhalents, nicotine
  • the therapeutically effective amount of a compound of Formula (I) is in a sufficient amount to ameliorate or prevent 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 preventing said disease or condition.
  • T-cell mediated hypersensitivity disease comprising administering to a subject a pharmaceutically 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 treating or preventing 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 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 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 preventing drug and/or alcohol addiction comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
  • Still another embodiment described herein relates to a method of ameliorating and/or preventing obesity, comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
  • Yet still another embodiment described herein relates to a method of ameliorating and/or preventing impaired cognition and/or a memory disorder comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
  • One embodiment described herein relates to a method of improving cognition or memory in a subject comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I)
  • Another embodiment described herein relates to a method of ameliorating and/or preventing inflammation due to an inflammatory disease comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
  • inflammatory diseases include rheumatoid arthritis, asthma, and psoriasis.
  • Some embodiment 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 CB 1 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 pharmaceutically 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 ID 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 ID 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 ID 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 ID NO: 2.
  • Another embodiment disclosed herein relates to a method for identifying a compound which binds to a cannabinoid receptor that includes labeling a compound of Formula I with a detectable label; and determining the number of occupied cannabinoid receptors.
  • the detectable label can be a radiolabel (e.g, [ 3 H]).
  • Any of the embodiments listed herein may further include identifying a subject in need of treatment or ameliorating of any disease or condition identified herein.
  • 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.
  • 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.
  • physiologically acceptable defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
  • 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).
  • PVP polyvinylpyrrolidone
  • 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 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.
  • 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.
  • 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.
  • AU formulations for oral administration should be in dosages suitable for such administration.
  • 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.
  • 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.
  • 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.
  • a suitable human dosage can be inferred from ED 50 or ID 50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals. [0187] 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.
  • 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.
  • Procedure 1 The analysis was performed on a combined prep/analytical Waters/Micromass system consisting of a ZMD single quadropole mass spectrometer equipped with electro-spray ionization interface.
  • the HPLC system consisted of a Waters 600 gradient pump with on-line degassing, a 2700 sample manager and a 996 PDA detector.
  • GC method 50 was used. Method 50 starts at 5O 0 C and has a gradient of 20 °C/min until 250 0 C then holds the temperature for 5 minutes.
  • the analysis was performed on an Aglient 6850 series GC system with capillary S/SL inlet and FID with EPC installation.
  • the column was a 30 m X 0.32 mm x 0.25 ⁇ m HP5 column.
  • CDI (4.53 g, 29 mmol, 4 eq) was added to 3-Amino-4-(2-carboxy- phenylsulfanyl)-benzoic acid (2.1 g, 7.3 mmol) dissolved in THF (30 ml). The reaction was stirred for 16h at room temperature. Water (200 ml) was then added to the mixture resulting in, after filtration and drying, 1.78g (91%) of the titled compound as a off-white solid.
  • Example 12 (ll-chloro-dibenzorb,fl rh41thiazepin-8-yl)- [2,4-dimethyl-phenylVpiperazin-
  • Example 13 1 l-chloro-dibenzorb,f
  • Example 14 4-[Yl l-chloro-dibenzo
  • Example 16 [ " 4-(2,4-Dimethyl-phenyl)-piperazin-l -Vl]-(I l-pentyl-dibenzo[b,f
  • Example 17 r4-(2,4-Dimethyl-phenyl)-piperazin- 1 -yll-( 11 -isobutyl-dibenzo [b,f] ⁇ ,41thiazepin-8-yl) methanone.
  • Example 18 (ll-Cvclohexyl-dibenzo[b,f1 ri,41thiazepin-8-ylV [4-(2.4-dimethyl-phenylV piperazin-l-yll methanone.
  • Example 19 Hl -f4-chloro-phenyiydibenzorb.fi [L41thiazepin-8-ylVl-r4-(2,4-dimethyl- phenyD-piperzin- 1 -yli-methanone.
  • Example 22 1 l-Pentyl-dibenzorb,f] ri,4 "
  • Example 23 l l-Isobutyl-dibenzo
  • Example 24 l l-Cyclohexyl-dibenzo
  • Example 25 4-[ ⁇ l l-Propyl-dibenzo
  • Example 26 4-
  • Example 27 4-
  • Example 28 4-[(l l-Isobutyl-dibenzo[b,f1 ri ⁇ lthiazepine- ⁇ -carbonylVaminol-piperidine-l- carboxylic acid ethyl ester.
  • Example 29 4-Ff 11 -Cvclohexyl-dibenzorb.fi ri,41thiazepine-8-carbonyl)-aminol- piperidine-1-carboxylic acid ethyl ester.
  • Example 30 4-[(l l-(4-chloro-phenyiydibenzo[b,fl [1.41thiazepine-8-carbonyl)-amino "
  • Example 30b Alternative synthesis of 4-[(l l-(4-chloro-phenyl)-dibenzo[ " b,f
  • Example 36 l l-(4-morpholinylVdibenzorb,f
  • Example 38 11 -(4-methylpiperazinyl)-dibenzo[ " b,f] ri,41thiazepin-8-carboxylic acid piperidin- 1 -ylamide
  • reaction mixture was concentrated in vacuo and purified by column chromatography using EtOAc (0-10%) in heptane as the eluent furnishing 338 mg of 4-(2-Methoxycarbonyl-benzyl)-3-nitro-benzoic acid ethyl ester as a colorless solid (1.13 mmol, 65%).
  • Example 50 1 l-Chloro-dibenzofTxflfMioxazepine- ⁇ -carboxylic acid pi ⁇ eridin-1-ylamide
  • Example 52 1 l-Phenyl-dibenzorb,fjri,41oxazepine-8-carboxylic acid piperidin-1 -ylamide
  • Example 54 ll-(4-Chlorophenyl)-dibenzo[&,/1[l,41oxazepine-8-carboxylic acid piperidin- 1-ylamide
  • Example 55 l l-Q-Chlorophenyiydibenzol ' ⁇ ./if ' l ⁇ ' loxazepine- ⁇ -carboxylic acid piperidin- 1-ylamide
  • Examples 61-66 are examples of compounds synthesised from S-bromo-l l-chloro-dibenzoffr/Jtl ⁇ thiazepine according to the general procedure for palladium catalysed Negishi couplings and the procedures described by Pandya et al. J. Org. Chem. (2003), 68, 8274-8276 and Sezen and Sanies et al., Org. Lett. (2003), 5, 3607-3610, which are both incorporated by reference in their entireties.
  • Example 62 11 -(4-ChlorophenylVdibenzo[6, f ⁇ ⁇ 1 ,4]thiazepine-8-sulfonic acid butylamide
  • Example 70 2-Fluoro-l l-oxo-10,1 l-dihydro-dibenzo[6,/][l,41thiazepine-8-carboxylic acid
  • Example 72 ll-Chloro-2-fluoro-dibenzo[6,/1[ " l,4]thiazepine-8-carboxylic acid (2- phenylpropylVamide
  • Example 73 l l-Chloro- ⁇ -fluoro-dibenzof ⁇ Jiri ⁇ jthiazepine- ⁇ -carboxylic acid (3- chlorobenzyD-amide
  • Example 74 ll-(4-Chlorophenyl)-2-fluoro-dibenzo[ ⁇ ,/][l,41thiazepine-8-carboxylic acid ⁇ -phenylpropylVamide
  • Example 75 1 l-(3-Chlorophenyl " )-2-fluoro-dibenzo[6,/1[l,4]thiazepine-8-carboxylic acid (2-phenylpropylVamide
  • Example 76 2-Fluoro-l l-piperidin-l-yl-dibenzo
  • Example 77 ll-(3-Chlorophenyl)-2-fluoro-dibenzo
  • Example 78 11 -(4-Chlorophenyl)-2-fluoro-dibenzo
  • Trifluoroacetic acid (90 rnL) was added to a solution of 4-tert- butylsulfanyl-3-nitrobenzoic acid ethyl ester (4.65 g; 16.4 mmol) in 20 mL dichloromethane. The mixture was stirred for 3 days at room temperature before evaporation of the solvent.
  • Example 111 1 l-Chloro-3-chloro-dibenzo[&,/][l,41thiazepine-8-carboxylic acid butyl amide
  • Example 112 l l-(4-Chlorophenyl)-3-chloro-dibenzo
  • Example 113 N-( 4-FluorobenzvD- l l-C4-chlorophenyr)-5-oxo-5H-5 ⁇ 4 - dibenzo ⁇ b,f ⁇ .41thiazepine-8-carboxamide
  • N-(4-Fluoroben2yl)-l l-(4-chlorophenyl)-dibenzo[&,/][l,4]thiazepine-8- carboxamide (182 mg, 0.385 mmol) was suspended in acetic acid (25 mL). Hydrogen peroxide (35% aqueous solution: 1.65 mL) was added dropwise to the suspension at room temperature. After ⁇ 5 hours stirring at room temperature the reaction mixture became clear yellow solution. The stirring was continued overnight at room temperature. The reaction mixture was slowly poured into saturated aqueous sodium bicarbonate (150 mL) - vigorous gas liberation. The neutralized mixture (pH ⁇ 7) was extracted with DCM.
  • the desired compound was isolated from the crude mixture, which was obtained during the preparation of N-(3-chlorobenzyl)-ll-(4-fluorophenyl)-5-oxo-5H-5 ⁇ 4 - dibenzo[ ⁇ j][l,4]thiazepine-8-carboxarnide. Purification by silica gel column chromatography eluting with a stepwise gradient of 20-50% ethyl acetate in heptane, afforded the desired compound (2.3 mg).
  • N-Butyl-11 -(4-chlorophenyl)-dibenzo[fr/J [1 ,4]thiazepine-8-carboxamide (70 mg; 0.17 mmol) was dissolved in DCM (10 niL) and 3-chloroperbenzoic acid (225 mg; 1.0 mmol) was added. After 4 hours stirring at room temperature the mixture was diluted with DCM (20 mL) and washed with saturated aqueous sodium hydrogen carbonate (3 x 15 mL). The organic phase was dried over sodium sulphate, filtered and evaporated to dryness. Purification by preparative TLC eluting twice with 50% ethyl acetate in heptane afforded the title compound (7.9 mg; 10 %).
  • Example 119 l l-(l-Oxy-piperidin-l-yl)-dibenzo[6,/]ri,41thiazepine-8-carboxylic acid 3- chlorobenzylamide (A) and 5-oxo-l l-piperidin-l-yl-5H-5 ⁇ 4 -dibenzo[6,/]-
  • Example 121 1 l-Cvclohexyl-5,5-dioxo-5H-5 ⁇ 4 -dibenzo[6,/][ " l,41thiazepine-8-carboxylic acid 4-fluorobenzylamide
  • the reaction mixture was heated in a sealed flask for 20 minutes at 170° C under microwave irradiation.
  • the reaction mixture was partitioned between DCM and weak acidic aqueous layer (10 mL water was acidified with 2-3 drops of concentrated HCl).
  • the organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue using a silica gel column chromatography, eluting with a stepwise gradient of 20 to 50% ethyl acetate in «-heptane, afforded the title compound (16 mg, 30%).
  • Ry 0.19 (EtOAc/n- ⁇ eptane 50:50).
  • Example 125 1 l-(4-Fluorophenyl)-N-(l-phenylethylV5H-dibenzo
  • Example 128 ir8-Chloro-l l-(4-fluorophenyl)-dibenzor6,eiri,41diazepin-5-yllethanone
  • N,N-Dimethyl amine (40 mg, 0.33 mmol) was added to a solution of 8- chloro-l l-(4-fluorophenyl)-5H-dibenzo[ ⁇ ,e][l,4]diazepine (108 mg, 0.33 mmol) in dry T ⁇ F (2 mL) at room temperature, followed by addition of acetyl chloride (70 ⁇ L, 0.99 mmol). The reaction mixture was shaken overnight at 60 0 C, allowed to cool to room temperature and partitioned between ethyl acetate and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness.
  • the crude mixture was passed over a short silica gel column using a mixture of ethyl acetate and n-heptane (30:70) as the eluant.
  • the isolated fractions were a mixture of the desired compound and a side product. The fractions were left on standing over the weekend.
  • the desired compound was crystallized in the fractions and it was isolated by filtration (69 mg, 60%).
  • R/ 0.20 (EtOAc/n-Heptane 50:50).
  • LCMS m/z 365 [M+H] + .. HPLC r R 5.0 min.
  • amidoimidoyl chlorides (Examples 147 - 162) were synthesized according to the general procedure for amide formation at 0.5 mmol scale except that the reaction mixture was passed through a pad of acidic alumina oxide and eluted with a mixture Of CH 2 Cl 2 and EtOAc. The eluents were concentrated at reduced pressure and the obtained crude products were directly used in the next reactions without further purifications or characterization.
  • Example 147 1 l-(chloro)-dibenzo[b,f][l,4]thiazepin-8-yl-(piperidin-l-yl)-methanone
  • Example 150 N-(butyl)-l l-(chloro)-dibenzorb,f1[l,41thiazepine-8-carboxamide
  • Example 151 N-(3-phenylpropyl)-l l-(chloroVdibenzo[b,f][l,4]thiazepine-8-carboxamide
  • Example 152 N-(2-phenylethyl)-l l-(chloro)-dibenzorb,f][l,4]thiazepine-8-carboxamide
  • Example 154 N-(2.4-dichlorobenzvD-l l-rchloroVdibenzorb.f
  • Example 156 N-(2-(3 -chlorophenvPethyD- 11 -(chloro)-dibenzo [b,f
  • Example 158 N-(2-bromobenzyl)-l l-(chloro)-dibenzo[b.f][l,4]thiazepine-8-carboxaniide
  • Example 160 N-((N-ethyl-N-phenyl)aminoethyl)-l l-rchloro)-dibenzo[b,f
  • Example 161 l l-rchloro)-dibenzo[b,f
  • Example 162 N-(4-fluorobenzyl)-l l-(chloro)-dibenzo[b.f
  • Example 163 1 l-(piperidinyl)-dibenzorb,firi,41thiazepin-8-yl-(piperidin-l-yl)-methanone
  • Example 170 N-(2,4-dichlorobenzyT)-l l-(piperidinyl)-dibenzo[b,f
  • Example 171 N-(2-(4-chlorophenyl)ethvD-l l-(piperidinylVdibenzo[b,fl
  • Example 172 N-(2-(3-chlorophenyl)ethyl)-l l-(piperidinyl)-dibenzo[b.fl[l,4]thiazepine-8- carboxamide
  • Example 175 N-( " 2-phenyl-propyl)-l l-fpiperidinylVdibenzorb.f
  • Example 176 N-( " (N-ethyl-N-phenyl)aminoethyl>-l l-(piperidinyl)- dibenzorb,firi,41thiazepine-8-carboxamide
  • Example 177 l l-(piperidinyl)-dibenzo[b,f
  • Example 178 N-(4-fluorobenzyl)-l l-(piperidinyl)-dibenzo[b,f
  • Example 179 1 l-fcvclohexyD-dibenzorb ⁇ firi ⁇ lthiazepin- ⁇ -yl-fpiperidin-l-vD-methanone
  • Example 190 N-((N-ethyl-N-phenyl)aminoethylVl l-(cyclohexyl)- dibenzorb.f] [1 ,41thiazepine-8-carboxamide
  • the arylzinc halide used for Examples 193 - 205 was 3-chlorophenylzinc iodide.
  • Example 193 1 l-(3-chlorophenyl)-dibenzo[b,f
  • Example 200 N-(2-(4-chlorophenyl)ethyl>l l-(3-chlorophenyl)-dibenzo[b,f1[L41thiazepine- 8-carboxamide
  • Example 201 N-(3-chlorobenzyl)-l l-(3-chlorophenyl)-dibenzo[b,f
  • Example 202 N-f ⁇ -phenyl-propyD-l l-O-chlorophenvD-dibenzorb ⁇ ri ⁇ lthiazepine-S- carboxamide
  • Example 204 l l-(3-chlorophenyl)-dibenzo[b,f
  • Example 205 N-(4-fluorobenzyl)-l l-(3-chlorophenyl)-dibenzo[b,f
  • Example 206 1 l-(4-fluorophenyl ' )-dibenzo[b,f
  • Example 212 N-(2,4-dichlorobenzyl)-l l-f4-fluorophenyl)-dibenzorb,firL41thiazepine-8- carboxamide
  • Example 216 N-(2-phenyl-propyl " )-l l-f4-fluorophenyl)-dibenzorb,f
  • Example 220 N-(I -phenylethy D-I l-(2-fluorophenyl)-dibenzo[b,f]fl,4]thiazepine- carboxamide
  • arylzinc halide used for Examples 233 - 246 was phenylzinc iodide.
  • Example 241 N-(2-(3-chlorophenyl)ethyl)-l l-fphenyl)-dibenzorb.fi ⁇ .41thiazepine- carboxamide
  • the arylzinc halide used for Examples 247 - 260 was 4-chlorophenylzinc iodide.
  • Example 247 1 l-(4-chlorophenyl)-dibenzo[b,f
  • Example 251 N-(3-phenylpropyl)-l l-f4-chlorophenyl)-dibenzo[b,fl[l ,4]thiazepine-8- carboxamide
  • Example 252 N-(2-phenylethyl)-l l-(4-chlorophenyl)-dibenzo[b,f
  • Example 258 N-(2-bromobenzyl)-l l-(4-chlorophenyl)-dibenzo[b,f
  • Example 260 N-((N-ethyl-N-phenyl)aminoethyl)-l l-r4-chlorophenyl)- dibenzo[b,f
  • Examples 272-301 are prepared according to the general procedure for the synthesis of amidines starting from 15 mg of the appropriate amidoimidoyl chloride (represented by titled compounds in Examples 262 - 271) and the appropriate amine (excess), except that purification is performed by eluting (EtOAc) the products through a pad of silica. The eluents are concentrated at reduced pressure to give the crude products, which are purified by preparative HPLC/MS. Yield is determined by weighing and purity by analytical LCVMS).
  • Examples 302-391 are prepared according to the general procedure for Negishi cross-coupling starting from 10-15 mg of the appropriate amidoimidoyl chloride (represented by titled compounds in Examples 262 - 271) and the proper arylzinc halide (8eq) in THF.
  • Ammonium chloride (0.02 ml) is added to the reaction mixtures, which are then passed through a short column (Na 2 SO 4 /silica) using EtOAc as eluent.
  • the eluents are concentrated at reduced pressure and the crude products are purified by preparative LC/MS. Yields are determined by weighing and purities by analytical LC/MS.

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

CB-I MODULATING COMPOUNDS AND THEIR USE
[0001] This application claims priority to U.S. Provisional Patent Application Serial Nos. 60/727,997, entitled "CB-I MODULATING COMPOUNDS AND THEIR USE", filed October 17, 2005; 60/831,003, entitled "CB-I MODULATING COMPOUNDS AND THEIR USE", filed July 14, 2006; 60/832,510, entitled "CB-I MODULATING COMPOUNDS AND THEIR USE", filed July 21, 2006; which are all incorporated by reference herein in their entireties, including any 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 CBl.
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 CBl 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). hi 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; Sanofi-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 CBl receptor antagonists and inverse agonists for the treatment of alcohol addiction, opiate addiction, cocaine addiction, anxiety, and septic shock. Interestingly, mice null for the CBl gene also display impaired cocaine 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 CB 1 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):
Figure imgf000004_0001
[0009] 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 and/or condition that would be alleviated, improved, and/or prevented by administration of a compound that modulates a cannabinoid receptor comprising administering to a therapeutically effective amount of a compound of Formula (I). Also disclosed herein are pharmaceutical compositions comprising a compound of Formula CD-
BRIEF DESCRIPTION OF THE DRAWINGS
[0010] Figure IA is a graph showing the percent response of the CBl receptor as the concentration of l l-Cyclohexyl-dibenzo[b,fj [l,4]thiazepine-8-carboxylic acid piperidin- 1-ylamide (Compound I) increases. Figure IB is a graph showing the percent response of the CB2 receptor as the concentration of Compound I increase.
[0011] Figure 2 is a bar graph showing the food intake in fasted rats 1 and 2 hours after being administered either 1, 3, or 10 mg/kg doses of Compound I. * Indicates p<0.05 as compared to the vehicle-treated controls. ** Indicates p<0.01 as compared to the vehicle- treated controls.
[0012] Figure 3 is bar graph showing the time course food intake in fasted rats after being administered 1 mg/kg of Compound I. * Indicates p<0.05 as compared to the vehicle-treated controls. ** Indicates p<0.01 as compared to the vehicle-treated controls.
[0013] Figure 4 is a bar graph showing cumulative food consumption at several points in time after the rats had been dosed with 10 mg/kg of Compound I. * Indicates p<0.05 as compared to the vehicle-treated controls.
[0014] Figure 5 A is a line graph showing the attenuation of CB 1 agonist-mediated effects after administration of CP 55,940 (0.3 and 1.0 mg/kg). Figure 5B is a line graph showing the attenuation of CBl agonist-mediated effects after administration of Compound I alone or in combination with CP55,940.
[0015] Figure 6 is a bar graph showing the body temperature of the rats at several points in time after the rats had been dosed with various doses of CP 55,950 or CP55,950 and Compound I.
[0016] Figure 7 is a bar graph showing the concentration of Compound I in the plasma and brain at several points in time.
[0017] Figures 8 A and 8B are bar graphs showing the concentration of compound, N-(butyl)- 11 -(4-chlorophenyl)-dibenzo [b,f,] [ 1 ,4]thiazepine-8 -carboxamide
(Compound II) in tissue and brain at several points in time. Figures 8C and 8D are line graphs showing the concentration of Compound II in the plasma and brain at several points in time.
[0018] Figure 9A in a line graph showing the effects of Compound II (1 and 3 mg/kg/day) on body weight Figure 9B is a line graph showing the effects of Compound II (1 and 3 mg/kg/day) on food intake and water intake. Figure 9C line graph showing the effects of Compound II (10 mg/kg/day) on body weight. Figure 9D is a line graph showing the effects of Compound II (10 mg/kg/day) on food intake and water intake. [0019] Figures 1OA and 1OC are bar graphs showing the exploration ratio at 1 and 2 hours after the mice had been dosed with the vehicle, CP 55,940 (0.3 mg/kg, ip), or SR 141716A (I mg/kg, ip). Figures 1OB and 1OD are bar graphs showing the discrimination index at 1 and 2 hours after the mice had been dosed with the vehicle, CP 55,940 (0.3 mg/kg, ip), or SR141716A (1 mg/kg, ip).
[0020] Figure 1 IA is a bar graph showing the exploration ratio 2 hours after the mice had been dosed with Compound II (3 mg/kg, ip). Figure 1 IB is a bar graph showing the discrimination index 2 hours after the mice had been dosed with Compound II (3 mg/kg, ip).
[0021] Figure 12 is a bar graph showing percentage of novel recognition of a familiar object 2 hours after the mice had been dosed with 1, 3, or 10 mg/kg of Compound II.
[0022] Figure 13 is a line graph showing the working memory errors of the mice after being dosed with the vehicle, tacrine (0.3 mg/kg), or Compound II (3 mg/kg).
[0023] Figure 14 is a line graph showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.05, 0.16, and 0.5 mg/kg).
[0024] Figure 15 is a line graph showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.05 mg/kg), Compound II (3.0 mg/kg), or apomorphine (0.05 mg/kg) and Compound II (3.0 mg/kg).
[0025] Figure 16 is a line graph showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.16 mg/kg), Compound II (3.0 mg/kg), or apomorphine (0.16 mg/kg) and Compound II (3.0 mg/kg).
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS [0026] One embodiment described herein relates to a compound of formula (I):
Figure imgf000006_0001
as a single isomer, a mixture of isomers, a racemic mixture of isomers, pharmaceutically acceptable salt, a solvate, metabolite or polymorph thereof, wherein: X can be selected from the group consisting of O, S, S=O, SO2, NRi, NC≡N, NC(=Z)Ri, NC(=Z)NRiaRib, CRlaRlb, C=O, C=CRiaRib, and SiRiaRib;
Y can be -N(R2) — or -C(RiR2) — ; the symbol ^7"= represents a single or double bond, where when =^= is a double bond, R2 is absent;
A can be selected from the group consisting of C3-C12alkyl, C4-Ci2alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, (cycloalkyl)alkyl, (cycloalkenyl)alkyl, (cycloalkynyl)alkyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, halogen, -NRiaR]b, -N=CR1 aRib, sulfenyl, sulfinyl, sulfonyl, and - (CH2)o-4-C(=Z)-ORi, wherein any member of said group can be substituted or unsubstituted;
B, C, D, E, F, G and I can be separately selected from the group consisting of 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)ORi, -C(=Z)NR,aRlb, -C(=Z)N(R,)NR,aR,b, -C(=Z)N(R1)N(R1)C(=Z)R1, -C(RO=NR1, -NRIaR,b, - N=CRlaRlb, -N(R,)-C(=Z)Ri, -N(R,)-C(=Z)NRlaRib, -S(O)NR,aR,b, -S(O)2NRlaRlb, -N(Ri)-S(=O)Ri, -N(Ri)-S(O)2R1, -0Rh -SRj, and -OC^Z)R1, wherein any member of said group can be substituted or unsubstituted except for hydrogen;
H can be selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, halogen, hydroxyl, nitro, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, -CN, -C(=Z)R,, -C(=Z)OR1; -C(=Z)NRlaRib, -Q=Z)N(R1)NR, ,R1 b, -C(=Z)N(Ri)N(R,)C(=Z)R1,
Figure imgf000007_0001
-NRlaRlb, -N=CRlaR,b, -N(R,)-C(=Z)R,, -N(RO-C(=Z)NRiaRlb, -S(0)NR,aRlb, -S(0)2NR,aRlb, -N(Ri)-S(=0)R,, -N(RO-S(O)2R1, - ORi, -SRi, and -OC(=Z)R!, wherein any member of said group can be substituted or unsubstituted;
Z can be O (oxygen) or S (sulfur);
Ri1 Ri a and R1 b can each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, -(CH2)0-7-OR3, - (CH2)O-7-SR3, -(CH2)o-7-NR3aR3b, haloalkyl, -C(=Z)R3, -C(=Z)OR3, and -C(=Z)NR3aR3b. wherein any member of said group can be substituted or unsubstituted except for hydrogen; or Rla and Rib 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 the group consisting of: 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;
R3, R3a, and R3b can each independently selected from the group consisting of: 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;
[0027] In some embodiments, A cannot be a substituted or unsubstituted piperazine.
[0028] In other embodiments, H cannot be selected from the group consisting of-
CF3, phenyl, -OS(O)2-CF3, methyl, -CN, halogen, and
Figure imgf000008_0001
when A is a substituted or unsubstituted heteroalicyclyl containing at least one nitrogen or-NRiaRib.
[0029] In still other embodiments, H cannot be halogen when A is substituted or unsubstituted aryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroaryl, halogen, and substituted or unsubstituted sulfenyl; X is -NR1, wherein Rj is hydrogen; and Y is -N(R2) =, wherein === is a double bond and R2 is absent.
[0030] In yet still other embodiments, when X is O or — NRi, wherein Ri is methyl and Y is -N(R2) =-=^= , wherein =^= is a double bond and R2 is absent then H cannot be - C(=Z)ORi, wherein Ri is hydrogen, methyl, or ethyl.
[0031] In one embodiments, when A is halogen, Y is -N(R2) ^=, wherein =^= is a double bond and R2 is absent, and X is S then F cannot be -S(O)2NRi aRib, wherein Riaand Rib are both hydrogen. [0032] In one embodiments, the compound of Formula (I) can bind to a cannabinoid receptor. In certain embodiments, the cannabinoid receptor can be a CBl receptor.
[0033] In some embodiments, Rj3 and R^ can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms and 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, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino. In other embodiments, R]3 and R^ can form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from the group consisting of:
Figure imgf000009_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, 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 the group consisting of:
Figure imgf000010_0001
[0034] In some embodiments, Rib can be hydrogen. In other embodiments, R^ can be C1-3alkyl.
[0035] In other embodiments, X can be S, SO, or SO2.
[0036] In one embodiment, H can be selected from the group consisting of aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, halogen, -C(=Z)Rj, -C(=Z)OR,, -C(=Z)NRlaRlb, -C(=Z)N(Ri)NRiaRib, -CC=Z)N(R1)N(R1)CC=Z)R1, - C(RO=NR1, -NRiaRib, -N=CRlaRlb, -N(Ri)-CC=Z)R1, -N(RO-CC=Z)NRi3RIb, -S(O)NRi8R15, -S(O)2NRl3Rlb, -N(Ri)-S(=O)Ri, -N(Ri)-S(O)2Ri, and -OC(=Z)Ri, wherein any member of said group can be substituted or unsubstituted. In another embodiment, H can be selected from the group consisting of cycloalkyl, cycloalkenyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, hydroxyl, sulfenyl, sulfinyl,' sulfonyl, haloalkoxy, -C(=Z)OR,, -C(=Z)N(Ri)NRi.Rib, -C(=Z)N(R,)N(Ri)C(=Z)R,, -C(Rj)=NR1, -NR,aR,b, - N=CRiaRib, -S(O)NRiaRib, -N(Ri)-S(O)Ri, -N(Ri)-S(O)2Ri, and -OC(=Z)Rb wherein any member of said group can be substituted or unsubstituted. In still another embodiment, H can be selected from the group consisting of cycloalkyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted. In yet still other embodiments, H can be an unsubstituted or substituted heteroaryl is selected from the group consisting of:
Figure imgf000010_0003
Figure imgf000010_0004
an
Figure imgf000010_0002
. In one embodiment, H can be an optionally substituted phenyl. In certain embodiments, the optionally substituted phenyl can be substituted with a Ci-4 alkyl.
[0037] In some embodiments, H can be -C(=Z)NRi3Rib In one embodiment, Rj8 can be selected from the group consisting of 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 certain embodiments, R1 a can be selected from the group consisting of alkyl, alkoxy, aryl, aralkyl, heteroaryl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted. In certain other embodiments, Ri a can an optionally substituted heteroaryl or heteroaralkyl. In some of the embodiments, wherein H can be -C(=Z)NRiaRib and alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH2)o-7-NR3aR3b then R^ can be hydrogen or methyl. In particular embodiments, the optionally substituted heteroaryl or heteroaralkyl can
be selectee I from the group consisting of:
Figure imgf000011_0001
Figure imgf000011_0002
embodiments, n can be 1 or 2. In more particular embodiments, the optionally substituted heteroaralkyl can be
Figure imgf000011_0003
, and in some embodiments, n can be 1 or 2.
[0038] In other embodiments, H can be -C(=Z)Rj or -C(=Z)ORi. In one embodiment, H can be -C(=Z)Ri and Ri can be selected from the group consisting of alkyl, cycloalkyl, aralkyl, halogen. In certain embodiments, H can be -C(=Z)ORi and Ri can be alkyl or aralkyl.
[0039] In still other embodiments, H can -C(=Z)N(R])N(Ri)C(=Z)Ri or -N(Ri)-C(=Z)NRlaRlb. In certain embodiments, -C(=Z)N(Ri)N(R1)C(=Z)Ri can be
Figure imgf000011_0004
wherein n is 0 or 1. In certain other embodiments, H can be -N(Rj)-C(^Z)NRi3RIb and Ri is hydrogen and Ri3 is alkyl or aralkyl. In any of the embodiments discussed in the present paragraph, R^ can be hydrogen.
[0040] In yet still other embodiments, H can be selected from the group consisting of -C(Ri)=NRi, -N(RO-CC=Z)Ri, and -OC(=Z)Ri. In certain enbodiments, H can be - C(Ri)=NRi, -N(Ri)-C(=Z)Ri, and -OC(=Z)Ri wherein at least on Rj is hydrogen or alkyl and at least one Ri is selected from the group consisting of alkyl, aryl, and aralkyl.
[0041] In some embodiments, H can be -N(Ri)-S(=O)Ri or -N(RO-SC=O)2Ri. In certain embodiments, H can be -N(Ri)-S(=O)Ri or -N(Ri)-S(K)^R1 and Ri can be hydrogen, aralkyl, or heteroaryl.
[0042] In other embodiments, H can be -S(O)NRiaRib or -S(O)2NRiaRib- In certain embodiments, H can be -S(O)NRiaRib or -S(O)2NRiaRib and Ria can be selected from the group consisting of alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl. In any of the embodiments discussed in the present paragraph, R^ can be hydrogen.
[0043] In one embodiments, H can be -S(O)NRi8RIb, -S(O)2NRiaRib, -C(=Z)NRiaRib or -C(=Z)N(Ri)NRiaRib and Ri, Rja and R^ can each independently selected from the group consisting of:
Figure imgf000013_0001
wherein: n can be 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 can be selected from the group consisting Of-N(R4)-, O and S;
R4 and R5 each 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, 0-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, sulfmyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino; and
R6, R6a, Rβb, Rβc and Red can 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, sulfmyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino; or wherein the substituents selected from the group consisting of R6, R68, R6b, Rβc, and R6d 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, R6a, R6b, R6C, and R6d. In certain embodiments discussed in this paragarph, H can be -C(=Z)NRiaRib. In certain embodiments discussed in this paragarph, H 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 certain embodiments discussed in this paragarph, H can be -C(=Z)NRlaRib and Rib can be hydrogen. In certain embodiments discussed in this paragarph, H can be -C(=Z)NRlaRib, Ri b can be hydrogen, and n can be 0, 1, or 2.
[0044] In some embodiments, R1, Ria, R2a, R2, R3, R3a> and R3b can be each independently selected from the group consisting of aryl, heteroaryl, heteroalicyclyl, aralkyl, heteraralkyl, or (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.
[0045] 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 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. In certain embodiments, A can be an aryl, heteroaryl, or heteroalicyclyl and is substituted with zero to five substituents, wherein each substituent can be independently selected from the group consisting of 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 certain embodiments when A is substituted phenyl, the phenyl can be substituted with a halogen, methoxy, or cyano group.
[0046] In some embodiments, X can be selected from the group consisting of S, S=O, and SO2; Y can be -N(R2) :r= or -C(R[R2) ==; the symbol = represents a single or double bond, where when =^= is a double bond, R2 is absent; A can be selected from the group consisting of C3-Ci2alkyl, C4-Ci2alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted; B, C, D, E, F, G and I can be separately selected from the group consisting of 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)ORi, -C(=Z)NRlaRlb, -C(=Z)N(R0NRlaRlb, -C(=Z)N(R,)N(RI)C(=Z)R1, - C(RO=NR1, -NRIaRIb, -N=CRlaR,b, -N(R,)-C(=Z)R,, -N(R,)-C(=Z)NRlaRlb, -S(O)NRlaRib, -S(O)2NRiaRib, -N(Ri)-S(K))Ri, -N(Ri)-S(=O)2Ri, -ORi, -SRh and -OC(=Z)R,, wherein any member of said group can be substituted or unsubstituted except for hydrogen; H can be selected from the group consisting of -C(=Z)NRlaRib, -C(=Z)N(Ri)NRiaRib, -C(=Z)N(Ri)N(Ri)C(=Z)Ri, and -C(Ri)=NRi, wherein any member of said group can be substituted or unsubstituted; Z can be O or S; Ri1 Ri3 and Rj t, can each independently selected from the group consisting of: 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 unsubstituted except for hydrogen; or Ri3 and Rib 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 the group consisting of: 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 R3, R3a, and R3b can each independently selected from the group consisting of: 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 the group consisting of 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 the group consisting of 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.
[0047] In some embodiments, A can be selected from the group consisting of C3- C12alkyl (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)-OR1. In other embodiments, A can be selected from the group consisting of 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)o-4-C(=Z)-ORi; and X can be S (sulfur). In certain embodiments, A can be - NRiaRib wherein Rja is an aryl (e.g.,optionally substituted phenyl) and R^ is hydrogen. In certain other embodiments, A can be -NRiaRjb wherein Rj3 is a phenyl group substituted with a halogen and R^ is hydrogen. In certain embodiments, A can be C3-C]2alkyl (e.g., n- propyl), C4-C12alkyl (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 certain embodiments, 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-, Cg-, Cio-, Cn-, C]2 alkyl. In other embodiments, A is not C4-, C5-, C6-, C7-, C8-, C9-, C10-, Cn-, Ci2 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, -NRiaRit,. In some embodiments, A is not -(CH2)o-4-C(=Z)-ORi .
[0048] In some embodiments, A can be selected from the group consisting of C3- C12alkyl, C4-Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and - (CH2)o-4-C(=Z)-OR1; X can be S (sulfur); andY can be -N(R2)- wherein the symbol — represents a double bond and R2 does not exist. In some embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRiaRib. In certain embodiments, A can be selected from the group consisting of C3-C]2alkyl, C4-C]2alkyl, 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 H 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 =r^= represents a double bond and R2 does not exist; and H can be -C(=Z)NRiaRib. In certain embodiments, A can be a cycloalkyl, a heteroalicyclyl, or -NRi3R-Ib group; X can be S; Y can be -N(R2) ^^ wherein the symbol =^ represents a double bond and R2 does not exist; and H 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 H can be -C(=Z)NR]aRib, wherein Rj3 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.
[0049] In some embodiments, A can be selected from the group consisting of C3- Ci2alkyl, C4-Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and - (CH2)o4-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 H can be -C(=Z)NRiaR1b, wherein Ri a can be an optionally substituted alkyl, alkoxy, or -(CH2)0-7-NR3aR3b. In other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRiaRib, wherein Rla can be an optionally substituted alkyl, alkoxy, or -(CH2)o-7-NR33R3b. In still other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRlaRib, wherein Rj3 can be an optionally substituted alkyl, alkoxy, or -(CH2)0-7-NR3aR3b- In yet still other embodiments, A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRiaRib group; X can be S; Y ean be -N(R2)=-= wherein the symbol = represents a double bond and R2 does not exist; and H can be -C(=Z)NRiaRib, wherein Ri3 can be an optionally substituted alkyl, alkoxy, or -(CH2)0-7- NR3aR3b- In certain embodiments, the alkyl can be Ci-6 alkyl. In certain other embodiments, the alkoxy is a Ci-6 alkoxy.
[0050] In some embodiments, A can be selected from the group consisting of C3- C^alkyl, C4-Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRjb, 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 H can be -C(=Z)NRiaRib, wherein Rj a is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In other embodiments, A can be selected from the group consisting of C3-Ci2alkyl, C4-Ci2alkyl, halogen, and -(CH2)o-4-C(=Z)- ORi; X can be S; Y can be -N(R2) = wherein the symbol :r^^ represents a double bond and R2 does not exist; and H can be -C(=Z)NRlaRib> wherein Riais an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In still other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRlaRib, wherein Riais an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In yet still other embodiments, A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRiaRib group; X can be S; Y ean be — N(R2)=^= wherein the symbol =^= represents a double bond and R2 does not exist; and H can be -C(=Z)NR]aRib> wherein Rla is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl. In certain embodiments, the optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl is selected from the group consisting of:
Figure imgf000019_0001
, and in some of the embodiments, n can be 1 or 2.
[0051] In some embodiments, A can be selected from the group consisting of C3- C12alkyl, 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 H can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted aryl or aralkyl. In other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted aryl or aralkyl. In still other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRlaR1b, wherein Rj3 is an optionally substituted aryl or aralkyl. In yet still other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRiaRib, wherein R]ais an optionally substituted aryl or aralkyl. In certain embodiments, the optionally substituted aryl or aralkyl can be selected from the group
consisting of:
Figure imgf000020_0001
,wherein Q can be -N(R4)-, oxygen or sulfur; and R4 can be hydrogen or C1-4alkyl, and in some of the embodiments, n can be 1 or 2.
[0052] In some embodiments, A can be selected from the group consisting of C3- C12alkyl, C4-C12alkyl, 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 H can be -C(=Z)NRlaRib, wherein Riais an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In other embodiments, A can be selected from the group consisting of C3-Ci2alkyl, C4-C12alkyl, 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 H can be -C(=Z)NRiaRib, wherein R1 a is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In still other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRlaR1b, wherein Rlais an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In yet still other embodiments, A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NRlaRib group; X can be S; Y ean be - N(R2) == wherein the symbol = represents a double bond and R2 does not exist; and H can be -C(=Z)NRiaRib> wherein Ri3 is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl. In certain embodiments, the optionally substituted heteroalicyclyl or
(heteroalicyclyl)alkyl can be selected from the group consisting of: ,
Figure imgf000021_0001
Figure imgf000021_0002
Figure imgf000021_0003
of the embodiments, n can be 1 or 2.
[0053] In some embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRlaRib, wherein Rlais an optionally substituted heteroaryl or heteroaralkyl. In other embodiments, A can be selected from the group consisting of C3-C12alkyl, 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 H can be -C(=Z)NRlaRib5 wherein Rla is an optionally substituted heteroaryl or heteroaralkyl. In still other embodiments, A can be selected from the group consisting of 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 H can be -C(=Z)NRiaRib, wherein Rj a is an optionally substituted heteroaryl or heteroaralkyl. In yet still other embodiments, A can be selected from the group consisting of cycloalkyl (e.g., cyclohexyl), a heteroalicyclyl (e.g., piperidine), or -NR)aRib group; X can be S; Y can be -N(R2) =^ wherein the symbol =^= represents a double bond and R2 does not exist; and H can be -C(=Z)NRiaRib, wherein Ria is an optionally substituted heteroaryl or heteroaralkyl. In certain embodiments, the optionally substituted heteroaralkyl is from the
group consisting of:
Figure imgf000022_0002
, and
Figure imgf000022_0001
, 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
Figure imgf000022_0003
, wherein Q can be oxygen or sulfur, and in some of the embodiments, n can be 1 or 2..
[0054] Another embodiment described herein relates to each of the compounds and formulae shown in the claims. In one embodiment, the compound of Formula (I) can be selected from the group consisting of:
Figure imgf000022_0004
[0055] Certain of the compounds of the present invention may exist as stereoisomers including optical isomers. The invention includes all stereoisomers and both the racerhic 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.
[0056] 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. [0057] Still another embodiment described herein relates to a pharmaceutical composition, comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier, diluent, or excipient.
Definitions
[0058] 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 patents, 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
[0059] . As used herein, any "R" group(s) such as, without limitation, R], Ri8 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 R3 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 imgf000023_0001
[0060] 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.
[0061] 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.
[0062] 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 mmore of the indicated substituents.
[0063] Unless otherwise indicated, when a substituent is deemed to be "optionally subsituted," or "substituted" it is meant that the subsitutent 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, sulfmyl, 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.
[0064] 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. [0065] 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 "Ci-C4 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 the group consisting of 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.
[0066] 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, 0-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.
[0067] 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. [0068] 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.
[0069] As used herein, "aryl" refers to a carbocyclic (all carbon) ring or two or more fused rings (rings that share two adjacent carbon atoms) that have a fully delocalized pi- electron system. 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.
[0070] As used herein, "heteroaryl" refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system), 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, sulfmyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof.
[0071] 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.
[0072] 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.
[0073] "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.
[0074] 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.
[0075] 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.
[0076] 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.
[0077] 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 aryloxyl and arylthio may be substituted or unsubstituted.
[0078] 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.
[0079] 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 Cio, 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.
[0080] 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 connetected 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.
[0081] 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.
[0082] 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 the group consisting of 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 quaternized; 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 the group consisting of 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 ate not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, morpholinyl, oxiranyl, piperidinyl iV-Oxide, piperidinyl, piperazinyl, pyrrolidinyl, 4-piperidonyl, pyrazolidinyl, 2-oxopyrrolidinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, and thiamorpholinyl sulfone.
[0083] 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.
[0084] 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.
[0085] 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.
[0086] As used herein, "halo" or "halogen" refers to F (fluoro), Cl (chloro), Br (bromo) or I (iodo).
[0087] 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, trifluoromethyl and l-chloro-2-fluoromethyl, 2- fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.
[0088] 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.
[0089] 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.
[0090] 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.
[0091] A "trihalomethanesulfonyl" group refers to an "X3CSO2-" group wherein X is a halogen.
[0092] A "cyano" group refers to a "-CN" group.
[0093] An "isocyanato" group refers to a "-NCO" group.
[0094] A "thiocyanato" group refers to a "-CNS" group.
[0095] An "isothiocyanato" group refers to an " -NCS" group. [0096] 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.
[0097] 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.
[0098] An "S-sulfonamido" group refers to a "-SO2NRARB" group in which RA and RB can be the same as defined with respect to O-carboxy. An S-sulfonamido may be substituted or unsubstituted.
[0099] An "N-sulfonamido" group refers to a "RSO2N(RA)-" group in which R and RA can be the same as defined with respect to O-carboxy. A sulfonyl may be substituted or unsubstituted.
[0100] 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.
[0101] An "O-carbamyl" group refers to a "-OC(=O)NRARB" group in which RA and RB can be the same as defined with respect to O-carboxy. An O-carbamyl may be substituted or unsubstituted.
[0102] An "N-carbamyl" group refers to an "ROC(=O)NRA -" group in which R and RA can be the same as defined with respect to O-carboxy. An N-carbamyl may be substituted or unsubstituted.
[0103] An "O-thiocarbamyl" group refers to a "-OC(=S)-NRARB" group in which RA and RB can be the same as defined with respect to O-carboxy. An O-thiocarbamyl may be substituted or unsubstituted.
[0104] An "N-thiocarbamyl" group refers to an "ROC(=S)NRA-" group in which R and RA can be the same as defined with respect to O-carboxy. An N-thiocarbamyl may be substituted or unsubstituted.
[0105] A "C-amido" group refers to a "-C(=O)NRARB" group in which RA and RB can be the same as defined with respect to O-carboxy. A C-amido may be substituted or unsubstituted. [0106] An "N-amido" group refers to a "RC(=O)NRA-" group in which R and RA can be the same as defined with respect to O-carboxy. An N-amido may be substituted or unsubstituted.
[0107] 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.
[0108] A lower aminoalkyl refers to an amino group connected via a lower alkylene group. A lower aminoalkyl may be substituted or unsubstituted.
[0109] A lower alkoxyalkyl refers to an alkoxy group connected via a lower alkylene group. A lower alkoxyalkyl may be substituted or unsubstituted.
[0110] 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).
[0111] Where the numbers of substituents 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 alkoxy groups containing one, two or three atoms.
[0112] 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)).
[0113] As employed herein, the following terms have their accepted meaning in the chemical literature.
AcOH acetic acid anhyd anhydrous
CDI 1 , 1 '-carbonyldiimidazole
DCM dichloromethane
DMF N, N-dimethy lformamide
DMSO dimethyl sulfoxide Et2O diethyl ether
EtOAc ethyl acetate
EtOH Ethanol
MeOH Methanol
NH4OAc ammonium acetate
Pd/C palladium on activated carbon
[0114] 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.
[0115] 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.
[0116] 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.
[0117] 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)
[0118] 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.
[0119] 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.
[0120] As used herein, to "detect" changes in the activity of CB 1 or of a CB 1 subtype 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.
[0121] An "agonist" is defined as a compound that increases the basal activity of a receptor (i.e. signal transduction mediated by the receptor).
[0122] 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.
[0123] 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.
[0124] 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 was 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.
[0125] 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.
[0126] As used herein, a "patient" refers to a subject that is being treated by a medical professional such as an M.D. or a D.V.M. 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.
[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 1-7. 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.
Figure imgf000037_0001
[0131] In Scheme 1, R1a, R1b, and A are as defined above for Formula I. Scheme 2
Figure imgf000037_0002
[0132] In Scheme 2, R1a and R1b are as defined above for Formula I. R3 and R4 can be selected from the same group of substituents as R1a and R1b as defined above for Formula I. Scheme 3
Figure imgf000038_0001
[0133] In Scheme 3, Rla, Rib, and A are as defined above for Formula I. R3 and R4 can be selected from the same group of substituents as R1^ and Rib as defined above for Formula I.
Scheme 4
Figure imgf000039_0001
[0134] In Scheme 4, Ria, Rib, and A are as defined above for Formula I. R3 and R4 can be selected from the same group of substituents as Rj3 and R^ as defined above for Formula I. Scheme 5
Figure imgf000039_0002
[0135] In Scheme 5, Ria, Rib, and A are as defined above for Formula I.
Figure imgf000040_0001
Figure imgf000040_0002
[0136] In Scheme 7, Ria, Rib, and A are as defined above for Formula I. R3 and R4 can be selected from the same group of substituents as Rj a and R^ as defined above for Formula I.
Methods of Use
[0137] 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.
[0138] One embodiment disclosed herein relates to a method of ameliorating or preventing 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 the group consisting of: a method of treating or preventing 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, cocaine, 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, 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 prevent 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 preventing said disease or condition.
[0139] 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, cocaine, 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, 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 pharmaceutically 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.
[0140] Some embodiments disclosed herein relate to a method for treating or preventing 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 a therapeutically effective amount of a compound of Formula I.
[0141] In certain embodiments, 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. [0142] In some embodiments, the disease or condition can be obesity, metabolic syndrome, appetite disorders, cachexia, high cholesterol, hyperlipidemia and/or diabetes.
[0143] In certain embodiments, the disease or condition can be of the gastrointestinal system such as emesis, nausea, gastric ulcers, diarrhea or intestinal disorders.
[0144] In some embodiments, the disease or disorder can be an inflammation disease (e.g., rheumatoid arthritis, asthma, psoriasis).
[0145] In certain embodiments, the disease or condition can be of the cardiovascular system such as hemorrhagic sock, septic shock, cirrhosis, atherosclerosis, and/or cardiovascular disorders.
[0146] In other embodiments, the disease or condition can be of the reproductive system such as infertility and/or premature abortion.
[0147] In some embodiments, the disease or condition can be of the visual system such as glaucoma, uveitis, retinopathy, dry eye and/or macular degeneration.
[0148] In certain embodiments, the disease or condition can be osteoporosis and/or ostepenia.
[0149] In other embodiments, the disease or condition can be asthma and/or pleurisy.
[0150] In certain embodiments, the disease or condition can be cancer.
[0151] Another embodiment described herein relates to a method of ameliorating and/or preventing drug and/or alcohol addiction comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
[0152] Still another embodiment described herein relates to a method of ameliorating and/or preventing obesity, comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
[0153] Yet still another embodiment described herein relates to a method of ameliorating and/or preventing impaired cognition and/or a memory disorder comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I).
[0154] One embodiment described herein relates to a method of improving cognition or memory in a subject comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I) [0155] Another embodiment described herein relates to a method of ameliorating and/or preventing inflammation due to an inflammatory disease comprising administering to a subject a pharmaceutically effective amount of a compound of Formula (I). A non-limiting list of inflammatory diseases include rheumatoid arthritis, asthma, and psoriasis.
[0156] Some embodiment 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 CB 1 receptor.
[0157] 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.
[0158] 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 pharmaceutically effective amount of a compound of Formula I.
' [0159] 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 ID 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 ID NO: 2.
[0160] 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 ID 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 ID NO: 2.
[0161] Another embodiment disclosed herein relates to a method for identifying a compound which binds to a cannabinoid receptor that includes labeling a compound of Formula I with a detectable label; and determining the number of occupied cannabinoid receptors. In one embodiment, the detectable label can be a radiolabel (e.g, [3H]).
[0162] Any of the embodiments listed herein may further include identifying a subject in need of treatment or ameliorating of any disease or condition identified herein.
[0163] 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.
Pharmaceutical Compositions
[0164] 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.
[0165] 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.
[0166] The term "physiologically acceptable" defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
[0167] 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.
[0168] 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.
[0169] 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.
[0170] 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.
[0171] 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.
[0172] 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.
[0173] 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.
[0174] 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. [0175] 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. AU formulations for oral administration should be in dosages suitable for such administration.
[0176] For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
[0177] 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.
[0178] 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.
[0179] 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.
[0180] Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[0181] 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.
[0182] 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.
[0183] 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.
[0184] 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.
[0185] 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.
[0186] 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 ID50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals. [0187] 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.
[0188] 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.
[0189] 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%.
[0190] In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
[0191] 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.
[0192] 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.
[0193] 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
[0194] 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 - General analytical LC-MS procedure
[0195] Procedure 1 (API): The analysis was performed on a combined prep/analytical Waters/Micromass system consisting of a ZMD single quadropole mass spectrometer equipped with electro-spray ionization interface. The HPLC system consisted of a Waters 600 gradient pump with on-line degassing, a 2700 sample manager and a 996 PDA detector.
[0196] Separation was performed on an X-Terra MS Cl 8, 5 μm 4.6x50mm column. Buffer A: 1OmM ammonium acetate in water, buffer B: 1OmM ammonium acetate in acetonitrile/water 95/5. A gradient was run from 30%B to 100%B in 10 min, dwelling at 100%B for 1 min, and re-equilibrating for 6 min. The system was operated at 1 ml/min. [0197] Procedure 2 (AP2): The analysis was performed on a combined prep/analytical Waters/Micromass system consisting of a ZMD single quadropole mass spectrometer equipped with electro-spray ionization interface. The HPLC system consisted of a Waters 600 gradient pump with on-line degassing, a 2700 sample manager and a 996 PDA detector.
[0198] Separation was performed on an X-Terra MS Cl 8, 5 μm 4.6x50mm column. Buffer A: 1OmM ammonium acetate in water, buffer B: 1OmM ammonium acetate in acetonitrile/water 95/5. A gradient was run from 30%B to 100%B in 7 min, dwelling at 100%B for 1 min, and re-equilibrating for 5.5 min. The system was operated at 1 ml/min.
Example 2 - General gas chromatography (GC) procedure
[0199] GC method 50 was used. Method 50 starts at 5O0C and has a gradient of 20 °C/min until 2500C then holds the temperature for 5 minutes. The analysis was performed on an Aglient 6850 series GC system with capillary S/SL inlet and FID with EPC installation. The column was a 30 m X 0.32 mm x 0.25 μm HP5 column.
Example 3: 4-("2-methoxycarbonyl-phenylsulfanyl)-3-nitro-benzoic acid ethyl ester
Figure imgf000054_0001
[0200] Methyl 2-mercaptobenzoate (4.67 ml, 34 mmol) was added during 30 min to a mixture of ethyl 4-flouro-3-nitrobenzoate (6.60 g, 30.9 mmol) and Cs2CO3 (10.06 g, 30.9 mol) in DMF (60 mL) at 40 0C. The reaction mixture was diluted with EtOAc, water after additional 15 min (full conversion according to TLC). The aqueous phase was extracted once with EtOAc and the combined organic phases were washed twice with water followed by brine and then dried (Na2SO4). Filtration and concentration of the organic phase at reduce pressure gave a yellow crystalline residue. Recrystallization from EtO Ac/heptane gave 10.3 g (92%) of the titled compound as yellow crystals. 1H NMR (400 MHz, CDCl3) δ 8.82 (d, IH, J = 1.9 Hz), 7.94 (m, 2H), 7.62-7.57 (m, 3H), 6.92 (d, IH, J = 8.6 Hz), 4.38 (q, 2H, J = 7.2 Hz), 3.78 (s, 3H), 1.38 (t, 3H, J = 7.0 Hz); 13C NMR (100 MHz, CDCl3); δ 166.8, 164.6, 145.5, 144.1, 137.6, 136.3, 133.4, 133.0, 131.5, 131.3, 130.5, 129.8, 128.1, 126.9, 61.9, 52.7, 14.5.
Example 4: 4-(2-carboxy-phenylsulfanyl)-3-nitro-benzoic acid
Figure imgf000055_0001
[0201] 4-(2-niethoxycarbonyl-phenylsulfanyl)-3-nitro-benzoic acid ethyl ester (9.56 g, 26.5 mmol) dissolved in THF (570 mL) and aqueous LiOH (264 ml, IM) was stirred at 60 0C for 2 h, then allowed to cool to room temperature. THF was removed at reduced pressure and the remaining aqueous mixture was extracted once with EtOAc. HCl (2M) was then added to the resulting aqueous solution until pH 2. The precipitation was filtred off, washed with water and finally dried, which afforded 8.7 g (99%) of the titled compound as yellow crystals. The crude product was sufficiently pure to be used in the next step without further purifications. 1H NMR (400 MHz, CD3OD) δ 8.71 (d, IH, J = 1.8 Hz), 7.95 (m, 2H), 7.64-7.59 (m, 3H), 7.00 (d, IH, J = 8.6 Hz); 13C NMR (100 MHz, CD3OD) δ 168.3, 166.1, 145.9, 143.3, 137.0, 136.5, 133.2, 132.6, 131.2, 131.1, 130.1, 130.0, 128.6, 126.3.
Example 5: 3-Amino-4-(2-carboxy-phenylsulfanylVbenzoic acid
Figure imgf000055_0002
[0202] Pd/C (10%, 200 mg) and PtO2 were added to 4-(2-carboxy- phenylsulfanyl)-3-nitro-benzoic acid (2.9 g, 9.1 mmol) dissolved in 100 ml of MeOH. The reaction flask were repeatedly evacuated and filled with H2. A balloon containing H2 was connected to the flask. After 16 h the reaction mixture was filtered through a pad of celite, which was then washed carefully with MeOH. Concentration of the filtrate at reduced pressure gave 2.5 g (96% yield, approximately 95% purity) of the titled compound as a white solid. The purity could be increased to 97% by recrystallization from EtOAc/MeOH (2.3g, 88% yield). 1H NMR (400 MHz, CD3OD) δ 8.01 (d, IH, J = 7.6 Hz), 7.51 (s, IH), 7.44 (d, IH, J = 8.0 Hz), 7.31 (d, IH, J = 8.0 Hz)5 7.28 (t, IH, J = 8.0 Hz), 7.16 (t, IH, J = 7.2 Hz), 6.74 (d, IH, J = 8.0 Hz); MS (ES+, M+l) = 290.
Example 6 : 11 -Oxo- 1 OJ 1 -dihvdro-dibenzo |"b,f| [" 1 ,4] thiazepine-8-carboxylic acid
Figure imgf000056_0001
[0203] CDI (4.53 g, 29 mmol, 4 eq) was added to 3-Amino-4-(2-carboxy- phenylsulfanyl)-benzoic acid (2.1 g, 7.3 mmol) dissolved in THF (30 ml). The reaction was stirred for 16h at room temperature. Water (200 ml) was then added to the mixture resulting in, after filtration and drying, 1.78g (91%) of the titled compound as a off-white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.78 (br s, IH), 7.77 (s, IH), 7.67 (m, 3H), 7.55-7.42 (m, 3H); 13C NMR (100 MHz, DMSO-d6); δ 168.9, 166.9, 140.3, 138.3, 136.0, 134.5, 133.5, 133.0, 132.9, 132.2, 132.1, 129.9, 126.5, 124.3.
Example 7: 11-Chloro-dibenzo fb,f|[l,41 thiazepine-8-carbonyl chloride
Figure imgf000056_0002
[0204] A solution of l l-Oxo-10,l l-dihydro-dibenzo [b,f][l,4] thiazepine-8- carboxylic acid_ (200 mg, 0.74 mmol) and phosphorus pentachloride (756 mg, 3.68mmol) in 4 mL toluene was heated to 110 0C for 2 h. Toluene and excess of phosphorus pentachloride was removed at reduced pressure to give the title compound (193 mg, 85%) as an yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.01 (d, IH, J = 2.0 Hz), 7.87 (dd, IH, J = 8.4, 2.2 Hz), 7.77 (m, IH), 7.58 (d, IH, J = 8.2 Hz), 7.47-7.44 (m, 2H), 7.44-7.39 (m, IH); 13C NMR (100 MHz, CDCl3); δ 167.5, 157.1, 146.7, 137.8, 137.4, 136.3, 134.5, 133.4, 133.3, 132.6, 130.3, 129.5, 129.1, 128.8; Example 7b: Alternative synthesis of 11-Chloro-dibenzo [b,f|[l,41 thiazepine-8-carbonyl chloride
[0205] A solution of SOCl2 (25 ml), l l-Oxo-10,l l-dihydro-dibenzo [b,f][l,4] thiazepine-8-carboxylic acid (1.24 g, 4.6 mmol) and DMF (0.05 ml) in toluene (25 ml) was heated at 80 0C for 17h. Toluene and excess SOCl2 were removed at reduced pressure to give 1.18 g (84%) of the title compounds as a yellow solid, which was used in the next step without further purifications. 1H NMR (400 MHz CDCl3) δ 8.01 (d, IH, j = 2.0 Hz), 7.87 (dd, IH, J = 8.4, 2.2 Hz), 7.77 (m, IH), 7.58 (d, IH, J = 8.2 Hz), 7.47-7.44 (m, 2H), 7.44-7.39 (m, IH); 13C NMR (100 MHz, CDCl3); δ 167.5, 157.1, 146.7, 137.8, 137.4, 136.3, 134.5, 133.4, 133.3, 130.3, 129.5, 129.1, 128.8.
Example 8: N-(butvD-l l-(chloro)-dibenzofb,f,]fl,4]thiazepine-8-carboxamide
Figure imgf000057_0001
[0206] 11-Chloro-dibenzo [b,fj[l,4] thiazepine-8-carbonyl chloride (616 mg; 2 mmol) dissolved in dry DCM (5mL) was added to a solution of butylamine (366 mg; 5 mmol) in dry DCM (1OmL) was added at 0 °C. The reaction was stirred for 30 min and then diluted with EtOAc. The organic phase was washed with NH4Cl (aq), brine and dried (Na2SO4). Filtration and evaporation at reduced pressure followed by purification by column chromatography (ethyl acetate/heptane 1:1) gave the title compound (557 mg, 81%) as a yellow solid. MS (ES+, M+l) = 345.
Example 9: N-(butyl)-l l-f4-chlorophenyl)-dibenzo[b,f,][l,41thiazepine-8-carboxamide
Figure imgf000057_0002
[0207] 4-Chlorophenylzinc iodide (0.5M in THF, 35mL) was added to N-(butyl)- l l-(chloro)-dibenzo[b,f,][l,4]thiazepine-8-carboxamide (2.8 g; 8.1 mmol) and PdCl2(PPh3)2 (5 mol%, 275 mg) in diy THF (90 niL) at room temperature. After 3h saturated aqueous NH4Cl and EtOAc was added and the aqueous phase was extracted twice with EtOAc. The combined organic phases were washed with brine and then dried (Na2SO4). Filtration, concentration at reduced pressure of the organic phase followed by purification by column chromatography (heptane/EtOAc 3:1 to 1:1) and recrystallization from toluene gave 2.86 g (84%) of the title compound as pale yellow crystals, m.p. 217-219 0C. 1H NMR (400 MHz, CDCl3) δ 7.75 (m, 2H), 7.64 (d, IH, J = 1.2 Hz), 7.55 (dd, IH, J= 7.8, 1.2 Hz), 7.50 (m, 2H), 7.42 (m, 3H), 7.31 (dt, IH, J = 7.6, 1.2 Hz), 7.16 (dd, IH, J = 7.6, 1.4 Hz). 6.06 (br s, IH), 3.44 (q, 2H, J = 7.2 Hz). 1.58 (m, 2H), 1.40 (m, 2H, J = 7.4 Hz), 0.95 (t, 3H, J = 7.2 Hz); MS (ES+, M+l) = 421.
Example 10: 1 l-Chloro-dibenzo[Z>,/][l,41thiazepine-8-carboxylic acid isobutylamide
Figure imgf000058_0001
[0208] A solution of 11-chloro-dibenzo [b,f][l,4]thiazepine-8-carbonyl chloride (0.59 g; 1.92 mmol) in DCM (10 mL) was added to a solution of isobutylamine (0.38 mL; 3.84 mmol) in DCM (10 mL) at 00C under argon. The mixture was stirred at room temperature for 1A hour. The reaction mixture was diluted with DCM and NH4Cl (sat). The aqueous phase was extracted twice with DCM and the combined organic phases dried over Na2SO4. After filtration and concentration by evaporation, the residue was purified by silica gel column chromatography eluting with 10-20 % EtOAc in n-heptane. 0.51 g (77%) of the title compound was obtained as a white powder.
[0209] 1H NMR (400 MHz, CDCl3) δ 7.77 -^ 7.73 (m, IH, ArH), 7.63 (dd, IH, J= 2.0, 8.0, ArH), 7.56 (d, IH, J = 2.0, ArH), 7.51 (d, IH, J = 8.0, ArH), 7.47 - 7.37 (m, 3H, ArH), 6.07 (br s, IH, NH), 3.26 (dd, 2H, J= 6.1, 6.8, CH2/Bu), 1.86 (sept, IH, J= 6.6, CH,Bu), 0.96 (d, 6H, J= 6.6, 2 x CH3). Example 11: l l-(5-Chlorothiophen-2-yl)-dibenzo["6,/]fl,41thiazepine-8-carboxylic acid isobutylamide
Figure imgf000059_0001
[0210] 5-Chloro-2-thienyl zinc bromide (0.5 M in THF, 3.5 mL; 1.72 mmol) was added to a solution of 1 l-chloro-dibenzofbjfJfl^jthiazepine-S-carboxylic acid isobutylamide (0.15 g; 0.43 mmol) and bis(triphenylphosphine)palladium(II)chloride (30 mg; 0.043 mmol) in 4 mL dry THF at room temperature. The mixture was stirred overnight at room temperature. The reaction mixture was partitioned between EtOAc and NH4C1 (sat). The organic layer was dried over Na2SO4, filtered and evaporated to dryness. The mixture was purified by silica gel column chromatography (10-30% EtOAc in n-heptane) and repurified by prep HPLC to afford the title compound as a yellow solid (27 mg; 15 %).
[0211] 1H NMR (400 MHz, CDCl3) δ 7.60 - 7.34 (m, 7H, ArH), 6.94 (d, IH, J = 4.0, thiophenH), 6.89 (d, IH, J= 4.0, thiopheneH), 6.15 (br m, IH, NH)5 3.26 (dd, 2H, J = 6.4, 7.2, CH2Bu), 1-87 (m, IH3 CHe11), 0.96 (d, 6H, J= 6.8, 2 x CH3). 13C NMR (100 MHz, CDCl3) δ 166.8, 162.7, 148.5, 145.1, 140.5, 137.1, 136.2, 135.3, 133.0, 132.8, 132.1, 132.0, 131.9, 130.3, 128.4, 127.3, 124.7, 123.9, 47.6, 28.8, 20.4. MS (ES+, M+l) = 427.
General Procedure A - Amide Formation:
[0212] A flame-dried flask was charged under argon with 11-Chloro-dibenzo [b,f][l,4] thiazepine-8-carbonyl chloride (180 mg; 0.58 mmol) in 4 mL dry DCM and cooled to 0 C. The amine (1.45 mmol) was then slowly added and the reaction was allowed to reach room temperature and stirred for 30 min. The reaction was diluted with DCM and the organic phase was washed with NH4Cl (aq), brine and dried (Na2SO4). Filtration and evaporation at reduced pressure follwed by purification by column chromatography (ethyl acetate/heptane 1:1) gave the compounds listed as Examples 12 -14 (72-88 %) as off-white solids. Example 12: (ll-chloro-dibenzorb,fl rh41thiazepin-8-yl)- [2,4-dimethyl-phenylVpiperazin-
1-yli-methanone.
Figure imgf000060_0001
[0213] The reaction was performed according to the general procedure A, which gave 220 mg (82%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ 7.75 (m, IH), 7.51 (d, IH, J = 8.0 Hz), 7.47-7.44 (m, 2H), 7.44-7.39 (m, IH), 7.31, (d, IH, J = 1.8 Hz), 7.24 (dd, IH, J = 7.8, 1.8 Hz), 7.02 (br s, IH), 6.98 (br d, IH, J = 8.0 Hz), 6.89 (d, IH, J = 8.0 Hz), 3.88 (br s, 2H), 3.54 (br s, 2H), 2.85 (br s, 4H), 2.28 (s, 6H); 13C NMR (100 MHz, CDCl3) δ 169.0, 156.2, 148.5, 146.4, 138.4, 137.9, 137.6, 133.6, 133.3, 133.1, 132.9, 132.3, 132.1, 130.2, 129.5, 129.1, 127.4, 126.1, 124.3, 119.4, 31.1, 20.9, 17.8; MS (ES+, M) = 462.
Example 13: 1 l-chloro-dibenzorb,f| [~l,4]thiazepin-8-carboxylic acid piperidin-1-ylamide.
Figure imgf000060_0002
[0214] The reaction was performed according to the general procedure A, which gave 157 mg (72%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ 7.74 (m, IH), 7.59 (dd, IH, J = 8.0, 1.8 Hz), 7.54 (s, IH), 7.50 (d, IH, J = 8.2 Hz), 7.47-7.43 (m, 2H), 7.43- 7.39 (m, IH), 2.80 (br s, 4H), 1.74 (br s, 4H), 1.44 (br s, 2H); 13C NMR (100 MHz, CDCl3) δ 164.3, 156.6, 146.5, 138.5, 138.1, 135.8, 133.5, 133.4, 132.7, 131.8, 130.4, 129.4, 126.7, 124.2, 57.7, 32.4, 25.8; MS (ES+, M+l) = 372.
Example 14: 4-[Yl l-chloro-dibenzo|~b,f| fM1thiazepine-8-carbonyl)-amino'|-piperidine-l- carboxylic acid ethyl ester.
Figure imgf000061_0001
[0215] The reaction was performed according to the general procedure A, which gave 189 mg (88%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ 7.74 (m, IH), 7.61 (dd, IH, J = 8.2, 1.9 Hz), 7.56 (d, IH, J = 1.6Hz), 7.51 (d, IH, J = 8.2 Hz), 7.47-7.44 (m, 2H), 7.44-7.39 (m, IH), 6.00 (d, IH, J = 7.6 Hz), 4.12 (m, 5H), 2.94 (t, 2H, J = 11.9 Hz), 2.00 (m, 2H), 1.38 (m, 2H), 1.26 (dt, 3H, J = 7.2, 1.6 Hz); 13C NMR (100 MHz, CDCl3) δ 165.6, 156.3, 155.7, 146.3, 138.3, 137.8, 136.0, 133.2, 133.2, 132.4, 131.6, 130.2, 129.1, 126.2, 123.9, 61.7, 47.5, 43.0, 32.2, 14.9; MS (ES+, M+l) = 444.
General Procedure B- Iron-Catalyzed Alkyl-Imidoyl Chloride Cross-Coupling
[0216] A flame-dried flask was charged under argon with the imidoyl chloride (0.05 mmol), Fe(acac)3 (0.9 mg, 0.0025 mmol), THF (1 mL) and NMP (0.1 mL). A solution of alkylmagnesium halogen (2M in Et2O, 100 μL, 0.20 mmol) was slowly added to the resulting red solution, causing an immediate colour change to dark brown. The resulting mixture was stirred for 10 min, and the reaction was then carefully quenched with NH4Cl (aq) and diluted with Et2O. The organic phase was washed with brine, dried (Na2SO4), filtered and evaporated to give the crude product. Purification by column chromatography (ethyl acetate/heptane/MeOH 1 :1:0.05) gave the product (60-90%).
Example 15: (11 -Butyl-dibenzo rb,fj [1 ,41thiazepin-8-vD- r4-C2,4-Dimethyl-phenyl> piperazin-1-vH methanone.
Figure imgf000062_0001
[0217] The reaction was performed according to the general procedure B, which gave 18.7 mg (77%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ 7.45 (m, 2H), 7.40-7.32 (m, 3H), 7.23 (d, IH, J = 1.8 Hz), 7.08 (dd, IH, J = 8.0, 1.8 Hz), 7.02 (br s, IH), 6.98 (br d, IH, J = 8.0 Hz), 6.89 (d, IH, J = 8.0 Hz), 3.88 (br s, 2H), 3.58 (br s, 2H), 3.05- 2.75 (m, 6H), 2.29 (s, 6H), 1.7 (m, 2H), 1.5 (m, 2H), 0.95 (t, 3H, J = 7.4 Hz); 13C NMR (100 MHz, CDCl3) δ 174.6, 169.7, 149.1, 148.6, 140.0, 139.0, 137.0, 133.5, 132.9, 132.8, 132.1, 130.8, 130.6, 128.8, 127.9, 127.4, 123.8, 123.8, 119.4, 42.3, 29.6, 22.7, 20.9, 17.8, 14.2; MS (ES+, M+l) = 484.
Example 16: ["4-(2,4-Dimethyl-phenyl)-piperazin-l -Vl]-(I l-pentyl-dibenzo[b,f| [1 ,4]thiazepin-8-yl)methanone.
Figure imgf000062_0002
[0218] The reaction was performed according to the general procedure B, which gave 20.1 mg (81%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ. 7.46 (m, 2H), 7.40-7.32 (m, 3H), 7.23 (d, IH, J - 1.6 Hz), 7.08 (dd, IH, J = 8.0, 1.8 Hz), 7.02 (br s, IH), 6.98 (br d, IH, J = 8.0 Hz), 6.89 (d, IH, J = 8.0 Hz), 3.88 (br s, 2H), 3.58 (br s, 2H), 3.05- 2.75 (m, 6H), 2.29 (s, 6H), 1.7 (m, 2H), 1.5-1.2 (m, 4H), 0.95 (t, 3H, J = 7.0 Hz); 13C NMR (100 MHz, CDCl3) δ 174.9, 170.0, 149.3, 148.9, 140.3, 139.3, 137.3, 133.8, 133.1, 133.1, 132.4, 131.1, 130.9, 129.0, 128.2, 127.6, 124.1, 119.7, 42.7, 32.0, 27.3, 22.9, 21.2, 18.1, 14.5; MS (ES+, M+l) = 498. Example 17 : r4-(2,4-Dimethyl-phenyl)-piperazin- 1 -yll-( 11 -isobutyl-dibenzo [b,f] π,41thiazepin-8-yl) methanone.
Figure imgf000063_0001
[0219] The reaction was performed according to the general procedure B, which gave 17.3 mg (72%) of the titled compound. MS (ES+, M+l) = 484.
Example 18: (ll-Cvclohexyl-dibenzo[b,f1 ri,41thiazepin-8-ylV [4-(2.4-dimethyl-phenylV piperazin-l-yll methanone.
Figure imgf000063_0002
[0220] The reaction was performed according to the general procedure B, which gave 16.8 mg (66%) of the titled compound. MS (ES+, M+l) = 510
Example 19: Hl -f4-chloro-phenyiydibenzorb.fi [L41thiazepin-8-ylVl-r4-(2,4-dimethyl- phenyD-piperzin- 1 -yli-methanone.
Figure imgf000063_0003
[0221] The reaction was performed according to the general procedure B, which gave 16.2 mg (60%) of the titled compound. MS (ES+, M) = 538. Example 20: 1 l-Propyl-dibenzorb,f1 rL41thiazepine-8-carboxyric acid piperidin-1-ylamide.
Figure imgf000064_0001
[0222] The reaction was performed according to the general procedure B, which gave 15.3 mg (81%) of the titled compound. MS (ES+, M+l) = 380.
Example 21: 11-Butyl-dibenzorbyf] [l,41thiazeρine-8-carboxylic acid piperidin-1-ylamide.
Figure imgf000064_0002
[0223] The reaction was performed according to the general procedure B, which gave 15.8 mg (80%) of the titled compound. MS (ES+, M+l) = 394.
Example 22: 1 l-Pentyl-dibenzorb,f] ri,4"|thiazepine-8-carboxylic acid piperidin-1-ylamide.
Figure imgf000064_0003
[0224] The reaction was performed according to the general procedure B, which gave 16.1 mg (79%) of the titled compound. MS (ES+, M+l) = 408.
Example 23: l l-Isobutyl-dibenzo|~b,fj [1 ,4]thiazepine-8-carboxylic acid piperidin-1- ylamide.
Figure imgf000065_0001
[0225] The reaction was performed according to the general procedure B, which gave 16.2 mg (82%) of the titled compound. MS (ES+, M+l) = 394.
Example 24: l l-Cyclohexyl-dibenzo|"b,f| [l,41thiazepine-8-carboxylic acid piperidin-1- ylamide.
Figure imgf000065_0002
[0226] The reaction was performed according to the general procedure B, which gave 15.9 mg (76%) of the titled compound. MS (ES+, M+l) = 420.
Example 25: 4-[ϊl l-Propyl-dibenzo|"b,f| fL4]thiazepine-8-carbonyl)-amino1-piperidine-l- carboxylic acid ethyl ester.
Figure imgf000065_0003
[0227] The reaction was performed according to the general procedure B, which gave 19.7 mg (87%) of the titled compound. MS (ES+, M+l) = 452.
Example 26: 4-|Yll-Butyl-dibenzo[b.f] |"l,41thiazepine-8-carbonyl)-amino]-piperidine-l- carboxylic acid ethyl ester.
Figure imgf000066_0001
[0228] The reaction was performed according to the general procedure B, which gave 19.2 mg (83%) of the titled compound. 1H NMR (400 MHz, CD3OD) δ 7.45 (dd, IH5 J = 1.4, 0.8 Hz), 7.44-7.37 (m, 3H), 7.34-7.28 (m, 3H), 4.03 (q, 2H5 J = 7.1 Hz), 4.03 (m, 2H), 3.92 (m, IH), 3.00 (m, IH), 2.84 (br t, 2H, J = 11.9), 2.78 (m, IH), 1.80 (d, 2H, J = 12.5 Hz)5 1.52 (m, 2H), 1.37 (m, 4H)5 1.15 (t, 3H, J = 7.0 Hz), 0.83 (t, 3H, J = 7.4 Hz); MS (ES+, M+l) = 466.
Example 27: 4-|Yll-Pentyl-dibenzo[b,f] [l,41thiazepine-8-carbonviyammo1-piperidine-l- carboxylic acid ethyl ester.
Figure imgf000066_0002
[0229] The reaction was performed according to the general procedure B, which gave 20.1 mg (84%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ 7.46 (m, 4H), 7.39-7.32 (m, 3H), 5.89 (d, IH5 J = 7.6 Hz)5 4.12 (q, 2H, J = 7.0 Hz), 4.10 (m, 3H)5 2.92 (m, 4H)5 1.98 (d, 2H5 J = 11.9 Hz), 1.68 (m, 2H)5 1.39 (m, 6H)5 1.25 (t, 3H5 J = 7.1 Hz), 0.90 (t, 3H, J = 7.2 Hz); 13C NMR (100 MHz3 CDCl3) δ 174.6, 165.9, 155.4, 148.6, 139.6, 138.7, 135.3, 132.6, 132.0, 130.7, 128.6, 127.6, 123.9, 123.0, 61.4, 47.1, 42.7, 42.2, 32.0, 31.4, 26.8, 22.4, 14.6, 13.9; MS (ES+, M+l) = 480.
Example 28: 4-[(l l-Isobutyl-dibenzo[b,f1 riΛlthiazepine-δ-carbonylVaminol-piperidine-l- carboxylic acid ethyl ester.
Figure imgf000067_0001
[0230] The reaction was performed according to the general procedure B, which gave 17.3 mg (74%) of the titled compound. 1H NMR (400 MHz, CDCl3) δ 7.46 (m, 4H), 7.39-7.31 (m, 3H), 5.98 (d, IH, J = 7.8 Hz), 4.12 (q, 2H, J = 7.0 Hz), 4.10 (m, 3H), 3.03 (dd, IH, J = 14.1, 5.5 Hz), 2.85 (t, 2H, J = 13.7 Hz), 2.63 (dd, IH, J = 14.1, 9.0 Hz), 1.98 (m, 3H), 1.35 (m, 2H), 1.25 (t, 3H, J = 7.1 Hz), 1.08 (d, 3H, J = 6.5Hz), 1.03 (d, 3H, J = 6.5 Hz); 13C NMR (100 MHz, CDCl3) δ 174.2, 166.2, 155.7, 148.8, 139.8, 139.0, 135.5, 132.9, 132.8, 132.4, 131.0, 128.9, 128.1, 124.3, 123.4, 61.6, 51.7, 47.4, 43.0, 42.2, 32.3, 27.3, 23.4, 22.4, 14.9; MS (ES+, M+l) = 466.
Example 29: 4-Ff 11 -Cvclohexyl-dibenzorb.fi ri,41thiazepine-8-carbonyl)-aminol- piperidine-1-carboxylic acid ethyl ester.
Figure imgf000067_0002
[0231] The reaction was performed according to the general procedure B, which gave 21.3 mg (87%) of the titled compound. MS (ES+, M+l) = 492.
Example 30: 4-[(l l-(4-chloro-phenyiydibenzo[b,fl [1.41thiazepine-8-carbonyl)-amino"|- piperidine-1-carboxylic acid ethyl ester.
Figure imgf000067_0003
[0232] The reaction was performed according to the general procedure B, which gave 18.2 mg (70%) of the titled compound. MS (ES+, M) = 520.
Example 30b: Alternative synthesis of 4-[(l l-(4-chloro-phenyl)-dibenzo["b,f| [l,4]thiazepine- δ-carbonvD-aminol-piperidine-l-carboxylic acid ethyl ester.
[0233] 4- chlorophenylzinc iodide (0.5M in THF, 11.5 ml, 5.76 mmol) was added dropwise to 4-[(l l-chloro-dibenzo[b,fj [l,4]thiazepine-8-carbonyl)-amino]-piperidine-l- carboxylic acid ethyl ester (640 mg, 1.44 mmol), and PdCl2(PPh3)2 (59 mg, 0.14 mmol, 0.1 eq) in dry THF (15 ml) at room temperature. After 30 min saturated aqueous NH4Cl and EtOAc was added and the aqueous phase was extracted once with EtOAc. The combined organic phases were washed with water, brine and then dried (Na2SO4). Filtration, concentration at reduced pressure of the organic phase followed by purification of the crude product by column chromatography (Heptane-EtOAc-MeOH 1:1 :0.01) gave 730 mg (97 %) of the titled compound as yellow crystals. 1H NMR (400 MHz, acetone-d6) δ 7.82 (d, 2H, J = 8.8 Hz), 7.78 (d, IH, J = 2.0 Hz), 7.62 (m, 3H), 7.58-7.52 (m, 4H), 7.45 (dt, IH, J= 8.8, 1.4 Hz), 7.29 (dd, IH, J = 5.8, 1.6 Hz), 4.08 (m, 5H), 2.96 (m, 2H), 1.93 (m, 2H), 1.52 (m, 2H), 1.22 (t, 3H, 7.0 Hz); 13C NMR (100 MHz, acetone-d6) δ 167.8, 165.1, 155.1, 148.7, 140.4, 139.0, 136.9, 136.8, 136.6, 132.4, 132.0, 131.5, 131.3, 130.5, 128.9, 128.7, 124.9, 124.1, 60.8, 47.4, 42.9, 31.9, 14.3.
General Procedure C: Palladium catalyzed Negishi cross-coupling of imidoyl chlorides and arylzinc halides.
[0234] The arylzinc halide (3-5 eq) was added to the imidoyl chloride (10 mg) and PdCl2(PPh3)2 (10 mol%) in dry THF (1 ml) at room temperature. After 30 min saturated aqueous NH4Cl and EtOAc was added and the aqueous phase was extracted once with EtOAc. The combined organic phases were washed with water, brine and then dried (Na2SO4). Filtration, concentration at reduced pressure of the organic phase followed by purification of the crude product by column chromatography (Heptane-EtOAc 1:1) gave the product. Example 31: l l-phenyl-dibenzo["b,f| [l,4]thiazepin-8-carboxylic acid piperidin-1-ylamide
Figure imgf000069_0001
[0235] The reaction was performed according to the general procedure C using l l-chloro-dibenzo[b,f] [l,4]thiazepin-8-carboxylic acid piperidin-1-ylamide and phenylzinc iodide, which gave 4.9 mg of the titled compound. MS (ES+, M+l) = 414.
Example 32: l l-("2-cvanophenyl)-dibenzo['b,f| [1.41thiazepin-8-carboxylic acid piperidin-1- ylamide
Figure imgf000069_0002
[0236] The reaction was performed according to the general procedure C using l l-chloro-dibenzo[b,fj [l,4]thiazepin-8-carboxylic acid piperidin-l-ylamide and 2- cyanophenylzinc iodide, which gave 5.4 mgof the titled compound. MS (ES+, M+l) = 439.
Example 33: I l-r3-bromophenyl)-dibenzo[b,f] [1.41thiazepin-8-carboxylic acid piperidin- 1-ylamide
Figure imgf000069_0003
[0237] The reaction was performed according to the general procedure C using l l-chloro-dibenzo[b,fj [l,4]thiazepin-8-carboxylic acid ρiperidin-1-ylamide and 3- bromophenylzinc iodide, which gave 6.4 mg of the titled compound. MS (ES+, M+l) = 492. Example 34: 1 l-(4-chlorophenyl)-dibenzo[b,f] [l,4]thiazepin-8-carboxylic acid piperidin-1- ylamide
Figure imgf000070_0001
[0238] The reaction was performed according to the general procedure C using l l-chloro-dibenzo[b,f] [l,4]thiazepin-8-carboxylic acid piperidin-1-ylamide and A- chlorophenylzinc iodide, which gave 5.4 mg of the titled compound. MS (ES+, M+l) = 439.
General Procedure D: Synthesis of Amidines
[0239] Imidoyl chloride l l-chloro-dibenzo[b,fj [l,4]thiazepin-8-carboxylic acid piperidin-1-ylamide (5 mg, 0.013 mmol) was mixed with an excess of the appropriate amine in dry toluene. The reaction was shaken for 18 h at 80 degrees C. Concentration of the reaction mixture at reduced pressure gave a crude product, which was purified by column chromatography (ethyl acetate/heptane 1:1 to 3:1).
Example 35: I l-piperidinyl-dibenzo|"b,f1 ri,4"|thiazepin-8-carboxylic acid piperidin-1- ylamide
Figure imgf000070_0002
[0240] The reaction was performed according to the general procedure D using piperidine, which gave 2.8 mg of the titled compound. MS (ES+, M+l) = 421.
Example 36: l l-(4-morpholinylVdibenzorb,f| [l,4]thiazepin-8-carboxylic acid piperidin-1- ylamide
Figure imgf000071_0001
[0241] The reaction was performed according to the general procedure D using 7 mg (0.019 mmol) of the imidoyl chloride and morpholine, which gave 5.9 mg of the titled compound. MS (ES+, M+l) = 423.
Example 37: l l-(propylaminyl)-dibenzo[b,f| |"l,4]thiazepin-8-carboxylic acid piperidin-1- ylamide
Figure imgf000071_0002
[0242] The reaction was performed according to the general procedure D using propyl amine except for applying lower reaction temperature (50 degrees), which gave 2.6 mg of the titled compound. MS (ES+, M+l) = 395.
Example 38: 11 -(4-methylpiperazinyl)-dibenzo["b,f] ri,41thiazepin-8-carboxylic acid piperidin- 1 -ylamide
Figure imgf000071_0003
[0243] The reaction was performed according to the general procedure D using 10 mg of the imidoyl chloride and methylpiperazine, which gave 7.6 mg of the titled compound. MS (ES+, M+l) = 436. Example 39: l l-phenylaminyl-dibenzo|rb,f| [l,4]thiazepin-8-carboxylic acid piperidin-1- ylamide
Figure imgf000072_0001
[0244] The reaction was performed according to the general procedure D using piperidine, which gave 2.6 mg of the titled compound. MS (ES+, M+l) = 429.
Synthesis of Carbon Analogs
Example 40: 4-(2-Methoxycarbonyl-bertzyl)-3-nitro-benzoic acid ethyl ester
Figure imgf000072_0002
[0245] A solution of methyl 2-(bromomethyl)benzoate (261 mg, 1.14 mmol) and tetrakis(triphenylphosphine)palladium(0) (52 mg, 0.045 mmol) in DME (2 mL) under argon was stirred at room temperature for lOmin. 4-Ethoxycarbonyl-2-nitrophenylboronic acid (308 mg, 1.29 mmol) dissolved in DME/EtOH 2:1 (3 mL) was added followed by 2M aq. Na2CO3 (2 mL) and stirring was continued for 2h. The reaction mixture was concentrated in vacuo and purified by column chromatography using EtOAc (0-10%) in heptane as the eluent furnishing 338 mg of 4-(2-Methoxycarbonyl-benzyl)-3-nitro-benzoic acid ethyl ester as a colorless solid (1.13 mmol, 65%).
[0246] 1H NMR (400MHz, CDCl3): 8.58 (d, 2H), 8.06 (dd, IH), 8.02 (dd, 2H), 7.50 (dt, IH), 7.38 (dt, IH), 7.18 (d, IH), 7.06 (d, IH), 4.69 (s, 2H), 4.39 (q, 2H), 3.76 (s, 3H), 1.40 (t, 3H). Example 41 : 4-(2-Carboxy-benzyl)-3-nitro-benzoic acid
Figure imgf000073_0001
[0247] A solution of 4-(2-Methoxycarbonyl-benzyl)-3-nitro-benzoic acid ethyl ester (159 mg, 0.46 mmol) in THF (14mL) and IM aq. LiOH (4.6 mL, 4.6 mmol) was stirred at 60 0C for 2h, then allowed to cool to room temperature. THF was removed at reduced pressure and the resulting aqueous mixture was treated with 2M HCl until the pH was about 1. Filtration provided 93 mg (0.3 mmol, 67%) of 4-(2-Carboxy-ben2yl)-3-nitro-benzoic acid as a yellow solid.
[0248] 1H NMR (400MHz, CD3OD): 8.49 (d, IH), 8.06 (dd, IH), 8.02 (dd, IH), 7.53 (dt, IH)5 7.40 (dt, IH), 7.26 (d, IH)5 7.12 (d, IH), 4.69 (s, 2H).
Example 42: 3-Amino-4-f2-carboxy-benzyl)-benzoic acid
Figure imgf000073_0002
[0249] A solution of 4-(2-Carboxy-benzyl)-3-nitro-benzoic acid (79mg, 0.26mmol) in MeOH (3mL) containing PtO2 (6mg) and Pd/C (7mg) was stirred under a hydrogen atmosphere for 2h at room temperature. Filtration and concentration in vacuo provided 71mg (0.267mmol, 100%) of 3-Amino-4-(2-carboxy-benzyl)-benzoic acid as yellow oil.
[0250] 1U NMR (400MHz, CD3OD): 7.26 (dd, IH), 7.44-7.38 (m, 2H), 7.32-7.26 (m, 2H), 7.16 (d, IH)5 6.87 (d, IH), 4.29 (s, 2H).
Example 43 : 6-Oxo-6.11 -dihydro-SH-dibenzorKeiazepine-S-carboxylic acid
Figure imgf000074_0001
[0251] To a stirred solution of 3-Amino-4-(2-carboxy-benzyl)-benzoic acid (70mg, 0.26mmol) in THF (3mL) at room temperature was added carbonyldiimidazole (167mg, 1.03mmol) in small portions and stirring was continued. After 4h, 4M HCl (3mL) was added followed by water. Filtration and drying provided 51mg (0.2mmol, 78%) of 6- Oxo-6,l l-dihydro-5H-dibenzo[b,e]azepine-3-carboxylic acid as a colourless solid. The product was further purified by crystallation from 2-propanol.
[0252] 1H NMR (400MHz, DMSOd6): 10.58 (s, IH), 7.70-7.61 (m, 3H), 7.48- 7.30 (m, 4H), 3.95 (s, 2H).
Example 44: 6-chloro-l lH-dibenzo["b,e"|azepine-3-carboxylic acid piperidin-1-ylamide
Figure imgf000074_0002
[0253] A solution of 6-oxy-5,6-dihydro-l lH-dibenzo[b,e]azepine-3-carboxylic acid (45 mg, 0.18 mmol) and phosphorus pentachloride (187 mg, 0.9 mmol) in 2 mL toluene was heated to 90 0C for 6 h. Toluene and excess of phosphorus pentachloride were removed at reduced pressure to give 60mg of 6-chloro-l lH-dibenzo[ό,e]azepine-3-carbonyl chloride. 1-Aminopiperidine (0.078 ml, 0.7 mmol) dissolved in CH2Cl2 was added to the crude acid chloride dissolved in CH2Cl2 at room temperature. EtOAc and H2O were added to the reaction mixture after Ih. The H2O phase was extracted once with EtOAc and the combined organic phases were washed with saturated aqueous NaHCO3 and brine and dried (Na2SO4). Filtration and concentration at reduced pressure of the organic phase followed by purification of the crude product by column chromatography (heptane-EtOAc 1:1) gave 25 mg (40%) of 6-chloro-l lH-dibenzo[b,e]azepine-3-carboxylic acid piperidin-1-ylamide. 1H NMR (400 MHz, CDCl3) δ 7.81 (d, 2H, J = 7.4 Hz), 7.68 (dd, IH, J = 8.0, 1.8 Hz), 7.59 (s, IH), 7.47 (dt, IH, J-= 7.4, 1.2 Hz), 7.33 (t, IH, J= 7.6 Hz), 7.27 (t, IH, J = 7.4 Hz), 3.74 (s, 2H), 2.83 (m, 4H), 1.72 (m, 4H), 1.42 (m, 2H); MS (ES+, M+l) = 354.
Example 45: 6-cyclohexyl-l lH-dibenzoPxelazepine-S-carboxylic acid piperidin-1-ylamide
Figure imgf000075_0001
[0254] The reaction was performed according to the general procedure for iron- catalyzed alkyl-imidoyl chloride cross coupling using 25 mg of 6-chloro-l lH- dibenzo[b,e]azepine-3-carboxylic acid piperidin-1-ylamide and an excess (0.35 ml) of cyclohexylmagnesium chloride (2M). This gave 13.7 mg (49%) of 6-cyclohexyl-l IH- dibenzo[b,e]azepine-3-carboxylic acid piperidin-1-ylamide. MS (ES+, M+l) = 402; UV/MS purity 100/100.
Synthesis of Oxygen Analogs
Example 46: 4-(2-Methoxycarbonyl-phenoxy)-3-nitro-benzoic acid ethyl ester
Figure imgf000075_0002
[0255] To a stirred solution of ethyl 4-fluoro-3-nitrobenzoate (2.53g, 11.87mmol) in DMF (4OmL) containing Cs2CO3 (4.26g, 13.06mmol) at 100 0C was added drop wise methyl salicylate (1.69mL, 13.06 mol) dissolved in DMF (40 mL) over 2h. After 15min the reaction mixture was allowed to reach room temperature and then diluted with EtOAc (10OmL) and washed with water (2xl00mL). The aqueous layer was extracted with DCM (10OmL). Drying (MgSO4) of the combined organic layers followed by filtration, concentration in vacuo and purification by CC using EtOAc (0-40%) in heptane provided 3.75g (10.85mmol, 91%) of 4-(2-Methoxycarbonyl-phenoxy)-3-nitro-benzoic acid ethyl ester as a yellow solid.
[0256] 1H NMR (400MHz, CDCl3): 8.60 (d, IH), 8.04 (dt, 2H), 7.62 (dt, IH), 7.38 (dt, IH), 7.19 (dd, IH), 6.73 (d, IH), 4.37 (q, 2H), 3.71 (s, 3H), 1.38 (t, 3H).
Example 47: 4-(2-Carboxy-phenoxy)-3-nitro-benzoic acid
Figure imgf000076_0001
[0257] A solution of 4-(2-Methoxycarbonyl-phenoxy)-3-nitro-benzoic acid ethyl ester (3.68 mg, 10.65 mmol) in THF (200 niL) and 1 M aq. LiOH (100 rnL, 100 mmol) was stirred at 60 0C for 2h, then allowed to cool to room temperature. THF was removed at reduced pressure and the resulting aqueous mixture was treated with 2 M HCl until the pH was about 1. Filtration provided 2.75g (9.08mmol, 85%) of 4-(2-Carboxy-phenoxy)-3-nitro- benzoic acid as a pale yellow solid.
[0258] 1H NMR (400MHz, CD3OD): 8.53 (d, IH), 8.10 (dd, IH), 8.04 (dd, IH), 7.69 (dt, IH), 7.42 (dt, IH), 7.26 (dd, IH), 6.82 (d, IH).
Example 48: 3-Amino-4-(2-carboxy-phenoxy')-benzoic acid
Figure imgf000076_0002
[0259] A solution of 4-(2-Carboxy-phenoxy)-3-nitro-benzoic acid (2.75 g, 9.08 mmol) in MeOH (80 niL) containing PtO2 (59 mg) and Pd/C (211 mg) was stirred for 2h under a hydrogen atmosphere at room temperature. Filtration and concentration in vacuo provided 2.47g (9.05 mmol, 100%) of 3-Amino-4-(2-carboxy-phenoxy)-benzoic acid as a pale yellow solid.
[0260] 1H NMR (400 MHz, CD3OD): 7.89 (dd, IH), 7.54-7.47 (m, 2H), 7.31 (dt, IH), 7.21 (dt, IH), 6.97 (d, IH), 6.68 (d, IH). Example 49: 11 -Oxo-10, 11 -dihydro-dibenzofb.fjf 1 ^"loxazepine-S-carboxylic acid
Figure imgf000077_0001
[0261] To a stirred solution of 3-Amino-4-(2-carboxy-phenoxy)-benzoic acid (2.44 g, 0.26 mmol) in THF (100 mL) at room temperature was added carbonyldiimidazole (3.7 g, 22.8 mmol) in small portions and stirring was continued. After 4 h, 4 M HCl (100 mL) was added followed by cupious amounts of water. Filtration and drying followed by crystallization (2-propanol) provided 1.017 g (3.99 mmol, 45%) of 11 -Oxo-10,11 -dihydro- dibenzo[b,fJ[l,4]oxazepine-8-carboxylic acid as white crystals.
[0262] 1H NMR (400 MHz, DMSO-d6): 10.61(s, IH), 7.77-7..74 (m, 2H), 7.67 (dd, IH), 7.60 (dt, IH), 7.39 (d, IH), 7.34 (d, IH) 7.31 (dt, IH).
Example 50: 1 l-Chloro-dibenzofTxflfMioxazepine-δ-carboxylic acid piρeridin-1-ylamide
Figure imgf000077_0002
[0263] To a stirred solution of 11 -Oxo-10,11 -dihydro- dibenzo[b,fJ[l,4]oxazepine-8-carboxylic acid (476 mg, 1.86 mmol) in toluene (20 mL) and thionyl chloride (20 mL) was added DMF (0.5 mL) and stirring was continued at 80 0C for 19 h. The reaction mixture was concentrated in vacuo and re-dissolved in anhydruos DCM (20 mL) and added to a solution of 1-aminopiperidine (604 μL, 5.59 mmol) dissolved in DCM (20 mL) at 0 0C and stirring was continued for 2h. The resulting reaction mixture was concentrated in vacuo and purified by CC using EtOAc (0-70%) in heptane affording 353 mg (0.99 mmol, 53%) of ll-Chloro-dibenzofbjfJfMJoxazepine-S-carboxylic acid piperidin-1- ylamide as a pale yellow solid.
[0264] 1H NMR (400 MHz, CDCl3): 7.77-7.72 (m, 2H), 7.63 (s, IH)5 7.53 (dt, IH)3 7.22 (dt, IH), 7.18 (dd, IH), 2.92 (br s), 1.76 (br s), 1.43 (br s). Example 51: l l-Cvclohexyl-dibenzoP3,f|["l,41oxazepine-8-carboxylic acid piperidin-1- ylamide
Figure imgf000078_0001
[0265] To a flame dried flask loaded with Fe(acac)3 under argon was added sequentially l l-Chloro-dibenzofbjfjfl^joxazepine-δ-carboxylic acid piperidin-1 -ylamide (79mg, 0.22mmol) dissolved in dry THF, NMP (0.5mL) and a 2M etheral solution of cyclohexylmagnesium chloride (440μL, 0.88mmol) at -78 0C and the reaction micture was allowed to slowly reach ambient temperature. After additionally 2h sat aq NH4Cl (5mL) was added followed by EtOAc (1OmL). After separation of the layers, the aq layer was extracted with EtOAc (2xl0mL). The combined organic layers were dried (MgSO4), filtered, concentrated in vacuo and purified by CC using EtOAc (0-50%) in heptane as the eluent affording 89mg (0.22mmol, 100%) of the l l-Cyclohexyl-dibenzo[b,fJ[l,4]oxazepine-8- carboxylic acid piperidin-1 -ylamide as a grey solid.
[0266] 1H NMR (400MHz, CDCl3): 7.65 (br s, IH), 7.63 (br s, IH), 7.45-7.39 (m, 2H), 7.21 (dt, IH), 7.15 (dd, 2H), 3.10 (br s), 2.91 (tt), 1.97 (d), 1.85 (br s), 1.74 (d), 1.61 (dd), 1.50 (br s), 1.42-1.29 (m), 1.25 (br s), 0.89-0.85 (m).
Example 52: 1 l-Phenyl-dibenzorb,fjri,41oxazepine-8-carboxylic acid piperidin-1 -ylamide
Figure imgf000078_0002
[0267] The title compound was synthesised by the same procedure as for preparation of l l-cyclohexyl-dibenzo[Z>,/][l,4]oxazepine-8-carboxylic acid piperidin-1- ylamide using l l-chloro-dibenzo[δ,/][l,4]oxazepine-8-carboxylic acid piperidin-1 -ylamide (19 mg; 0.05 mmol), phenylmagnesium bromide (3M in diethyl ether; 100 μL; 0.3 mmol), Fe(acac)3 (3 mg) and NMP (50 μL) in 1 mL dry THF. The titled copound was purified by preparative HPLC. Yield: 5.3 mg. LCMS m/z 398 [M+H]+. HPLC tR = 7.76 min.
Example 53: ll-('4-FluorophenylVdibenzo['^,/][l,41oxazepine-8-carboxylic acid piperidin- 1-ylamide
Figure imgf000079_0001
[0268] The title compound was synthesised by the same procedure as for preparation of l l-cyclohexyl-dibenzo[δ,/][l,4]oxazepine-8-carboxylic acid piperidin-1- ylamide using l l-chloro-dibenzo[δ,/J[l,4]oxazepine-8-carboxylic acid piperidin-1-ylamide (19 mg; 0.05 mmol), 4-fluorophenylmagnesium bromide (2M in diethyl ether; 150 μL; 0.3 mmol), Fe(acac)3 (3 mg) and NMP (50 μL) in 1 mL dry THF. The titled copound was purified by preparative HPLC. Yield: 3.9 mg. LCMS m/z 416 [M+H]+. HPLC tR = 7.97 min.
Example 54: ll-(4-Chlorophenyl)-dibenzo[&,/1[l,41oxazepine-8-carboxylic acid piperidin- 1-ylamide
Figure imgf000079_0002
[0269] The title compound was synthesised by the same procedure as for preparation of l l-cyclohexyl-dibenzo[δ,/J[l,4]oxazepine-8-carboxylic acid piperidin-i- ylamide using l l-chloro-dibenzo[ό,/][l,4]oxazepine-8-carboxylic acid piperidin-1-ylamide (19 mg; 0.05 mmol), 4-chlorophenylmagnesium bromide (IM in diethyl ether; 300 μL; 0.3 mmol), Fe(acac)3 (3 mg) and NMP (50 μL) in 1 mL dry THF. The titled copound was purified by preparative HPLC. Yield: 2.1 mg. LCMS m/z 432 [M+H]+, 434 [M+2+H]+. HPLC tR = 8.63 min.
Example 55: l l-Q-Chlorophenyiydibenzol'ά./if''loxazepine-δ-carboxylic acid piperidin- 1-ylamide
Figure imgf000080_0001
[0270] The title compound was synthesised by the same procedure as for preparation of l l-cyclohexyl-dibenzo[δ,/][l,4]oxazepine-8-carboxylic acid piperidin-1- ylamide using l l-chloro-dibenzo^/jfl^oxazepine-δ-carboxylic acid piperidin-1-ylamide (19 mg; 0.05 mmol), 3-chlorophenylzinc iodide (0.5M in THF; 600 μL; 0.3 mmol) and PdCl2(Ph3P)2 (3 mg) in 1 niL dry THF. The titled copound was purified by preparative HPLC. Yield: 8.7 mg. LCMS m/z 432 [M+H]+, 434 [M+2+H]+. HPLC tR = 8.63 min.
Synthesis of 8-bromo analogs:
Example 56: 2-(4-Bromo-2-nitrophenylsulfanyl) benzoic acid methyl ester
Figure imgf000080_0002
[0271] 5-Bromo-2-fluoronitrobenzene (1.23 mL; 10.0 mmol) and Cs2CO3 (3.58 g; 11.0 mmol) was mixed in 30 mL DMF and heated to 700C. A solution of methyl 2- mercaptobenzoate (1.5 mL mg; 10.9 mmol) in 30 mL DMF was added dropwise over 15 min. The heating was turned of and the mixture left stirring overnight at room temperature. Water and ethyl acetate was added and the aqueous layer extracted twice with ethyl acetate/heptane. After separation of the phases, the organic phase was washed twice with water, before drying over sodium sulphate, filtration and concentration in vacuo. Purification was done by silica gel column chromatography (0-30 % ethyl acetate in heptane) to afford the title compound as a yellow solid (3.61 g; 98%).
[0272] 1H NMR (400 MHz, CDCl3) δ 8.30 (d, IH, J= 2.4), 7.95 - 7.92 (m, IH), 7.54 - 7.45 (m, 4H), 6.86 (d, IH, J= 8.4), 4.82 (s, 3H). HPLC tR = 4.97 min.
Example 57: 2-(4-Bromo-2-mtrophenylsulfanyl) benzoic acid
Figure imgf000081_0001
[0273] Ester 2-(4-bromo-2-nitrophenylsulfanyl) benzoic acid methyl ester (3.57 g; 9.7 mmol) was dissolved in 120 mL THF and IM LiOH (aq, 60 mL) added. The solution was heated to 70 °C and stirred at that temperature for 2 hours. The temperature was allowed to cool to room temperature over 3 hours and THF was removed at reduced pressure. The remaining aqueous mixture was extracted once with EtOAc/heptane (1:1, 75 mL). HCl (2M) was then added to the resulting aqueous solution until pH 2. The precipitates were collected by filtration, washed with water and finally dried, which afforded the title compound as a yellow solid (2.82 g; 82%) that was used without further purification.
[0274] 1H NMR (400 MHz, CD3OD) δ 8.59 (d, IH, J= 2.0), 8.02 (dd, IH, J = 2.0, 8.8), 7.94-7.90 (m, IH), 7.65-7.57 (m, 3H), 7.08 (d, IH, J= 8.8).
Example 58: 2-(2-Amino-4-bromophenylsulfanyl) benzoic acid
Figure imgf000081_0002
[0275J 2-(4-Bromo-2-nitrophenylsulfanyl) benzoic acid (1.1 g; 3.1 mmol) was dissolved in 100 mL absolute ethanol and a catalytic amount of palladium on activated carbon was added. The reaction flask was evacuated and equipped with a balloon containing hydrogen. This procedure was repeated twice before the mixture was left stirring overnight at room temperature. The reaction mixture was filtered through a pad of celite and the solvent removed by evaporation to give the crude product (930 mg; 93 %) that was used without further purification. LCMS m/z 324 [M+H]+, 326 [M+2+H] , HPLC tR = 10.28 min.
Example 59: 8-Bromo-10H-dibenzo[ά,/|[l,4"|thiazepin-l l-one
Figure imgf000082_0001
[0276] 2-(2-Amino-4-bromophenylsulfanyl) benzoic acid (930 mg; 2.87 mmol) was dissolved in 25 mL dry TΗF and CDI was added (1.4 g mg; 8.61 mmol). The mixture was stirred at room temperature for 2/4 days before addition of 4 M aqueous HCl and water. The title compound precipitates and was collected by filtration to afford the desired lactam as colourless crystals (4.45 g; 85 %). LCMS m/z 306 [M+Η]+, 308 [M+2+H] , HPLC tR= 3.87 min.
Example 60: 8-Bromo-l l-chloro-dibenzo[6./iri,41thiazepine
Figure imgf000082_0002
[0277] Lactam 8-bromo-10H-dibenzo[6,/J[l,4]thiazepin-l l-one (748 mg; 2.44 mmol) was mixed with thionyl chloride (18 mL) in toluene (18 mL). DMF was added (200 μL) and the mixture stirred for 3 hours. After cooling the solvents were removed by evaporation under reduced pressure. Purification was done by silica gel column chromatography (0 - 20 % ethyl acetate in heptane) to afford the imidoyl chloride as a white powder. LCMS m/z 324 [M+Η]+, 326 [M+2+H]+, 328 [M+4+H]+ , HPLC tκ = 6.00 min.
[0278] The following compounds (Examples 61-66) are examples of compounds synthesised from S-bromo-l l-chloro-dibenzoffr/Jtl^thiazepine according to the general procedure for palladium catalysed Negishi couplings and the procedures described by Pandya et al. J. Org. Chem. (2003), 68, 8274-8276 and Sezen and Sanies et al., Org. Lett. (2003), 5, 3607-3610, which are both incorporated by reference in their entireties.
Example 61 : 8-Bromo- 11 -f 4-chlorophenyl)-dibenzo \b, f] F 1 ,4"|thiazepine
Figure imgf000083_0001
Example 62: 11 -(4-ChlorophenylVdibenzo[6, f\ \1 ,4]thiazepine-8-sulfonic acid butylamide
Figure imgf000083_0002
Example 63: l l-(4-Chlorophenyl)-dibenzo[Z>,/][L41thiazepine-8-sulfonic acid piperidin-1- ylamide
Figure imgf000083_0003
Example 64: 1 l-(4-Chlorophenyl')-8-oxazol-2-yl-dibenzo['6,/iri,41thiazepine
Figure imgf000083_0004
Example 65: 1 l-(4-Chlorophenyl)-8-thiazol-2-yl-dibenzo[6,/]|"l,41thiazepine
Figure imgf000084_0001
Example 66; 1 l-(4-Chlorophenyl)-8-imidazol-2-yl-dibenzo|"ά,/||~h41thiazepine
Figure imgf000084_0002
Synthesis of 2-fluoro analogs:
Example 67: 4-(4-Fluoro-2-ethoxycarbonylphenylsulfanyl>3-nitrobenzoic acid ethyl ester
Figure imgf000084_0003
[0279] Ethyl 4-fluoro-3-nitrobenzoate (953 mg; 4.47 mmol) and Cs2CO3 (1.54 g; 4.72 mmol) were mixed in 20 mL DMF and heated to 800C. A solution of ethyl 5-fluoro-2- mercaptobenzoate (808 mg; 4.34 mmol) in 20 mL DMF was added dropwise over 15 min. The heating was turned of and the mixture left stirring overnight at room temperature. Water and ethyl acetate was added and the aqueous layer extracted twice with ethyl acetate/heptane. The combined organic phases were washed with water before drying over sodium sulphate, filtration and evaporation of the solvents. Purification was done by silica gel column chromatography (0-10 % THF in heptane) to afford the title compound as a yellow oil (1.55 g; 94%).
[0280] 1H NMR (400 MHz, CDCl3) δ 8.84 (d, IH, J= 2.0), 7.96 (dd, IH, J= 1.6, 8.4), 7.67 - 7.62 (m, 2H), 7.32 - 7.27 (m, IH), 6.87 (d, IH, J= 8.4), 4.38 (q, 2H, J = 7.2), 4.23 (q, 2H, J = 7.2), 1.38 (t, 3H, J = 7.2), 1.17 (t, 3H, J = 7.2). LCMS m/z 394 [M+H]+ , HPLC tκ = 5.43 min.
Example 68: 4-(4-Fluoro-2-carboxyphenylsulfanyl>3-nitrobenzoic acid
Figure imgf000085_0001
[0281] Diester 4-(4-fluoro-2-ethoxycarbonylphenylsulfanyl)-3-nitrobenzoic acid ethyl ester (1.45 g; 3.8 mmol) was dissolved in 100 mL THF and IM LiOH (aq, 30 mL) added. The solution was heated to 70 0C and stirred at that temperature for 4 hours. The temperature was allowed to cool to room temperature and THF was removed at reduced pressure. The remaining aqueous mixture was extracted once with EtOAc. HCl (2M) was then added to the resulting aqueous solution until pH 2. The precipitates were filtered off, washed with water and finally dried, which afforded the title compound (1.22 g; 99%).
[0282] 1H NMR (400 MHz, CD3OD) δ 8.77 (d, IH, J = 1.6), 8.00 (dd, IH, J = 2.0, 8.4), 7.78 - 7.73 (m, 2H), 7.45 (dt, IH3 J= 3.2, 8.4), 7.01 (d, IH, J= 8.8).
Example 69: 3-Amino-4-(4-fluoro-2-carboxyphenylsulfanyl)benzoic acid
Figure imgf000085_0002
[0283] Diacid ,4-(4-fluoro-2-carboxyphenylsulfanyl)-3-nitrobenzoic acid (728 mg; 2.16 mmol), was dissolved in 50 mL absolute ethanol and stannous chloride, dihydrate (2.43 g; 10.8 mmol) was added. The temperature was raised to 70 0C and the temperature attained for 15 min. The heating was turned of and the flask slowly allowed to reach room temperature. Water was added and the aqueous phase extracted with ethyl acetate (3 times). The combined organic phases were washed extensively with brine, before drying over sodium sulphate, filtration and removal of the solvent by evaporation. The crude product was obtained as a pale yellow powder (320 mg; 48 %) that was used without further purification.
[0284] 1H NMR (400 MHz, CD3OD) δ 7.70 (dd, IH, J= 2.8, 9.2), 7.50 (d, IH, J = 2.0), 7.44 (d, IH, J= 8.0), 7.33 - 7.29 (m, IH), 7.12 - 7.05 (m, IH), 6.74 (dd, IH, J= 4.8, 9.2).
Example 70: 2-Fluoro-l l-oxo-10,1 l-dihydro-dibenzo[6,/][l,41thiazepine-8-carboxylic acid
Figure imgf000086_0001
[0285] 3-Amino-4-(4-fluoro-2-carboxyphenylsulfanyl)benzoic acid (320 mg; 1.04 mmol) was dissolved in 10 mL dry THF and CDI was added (675 mg; 4.17 mmol). The mixture was stirred at room temperature for 21A days before addition of 4 M aqueous HCl and water. The title compound precipitates and was collected by filtration to afford the desired lactam as colourless crystals (199 mg; 66 %).
[0286] 1H NMR (400 MHz, DMSO-cfe) δ 7.82 - 7.75 (m, IH), 7.54 - 7.52 (m, 3H), 7.51 - 7.42 (m, IH), 7.40 - 7.29 (m, IH).
Example 71: 11 -Chloro-4-fluoro-dibenzo [b, f] [ 1 ,4")thiazepine-8-carbonyl chloride
Figure imgf000086_0002
[0287] The lactam 2-fluoro-l l-oxo-10,l l-dihydro-dibenzo[έ,/][l,4]thiazepine-8- carboxylic acid (199 mg; 0.69 mmol) was mixed with thionyl chloride (8 mL) in toluene (8 niL). DMF (100 μL) was added and the mixture stirred at 8O0C overnight. After cooling the solvents were removed by evaporation under reduced pressure to afford the crude, yellow dichloride as a powder that was used immediately without further purification.
Example 72: ll-Chloro-2-fluoro-dibenzo[6,/1["l,4]thiazepine-8-carboxylic acid (2- phenylpropylVamide
Figure imgf000087_0001
[0288] The title compound was synthesized by the general procedure for amide formation using l l-chloro-4-fluoro-dibenzo[δ,/J[l,4]thiazepine-8-carbonyl chloride (~ 0.35 mmol), 8 mL dry dichloromethane and 2-phenylpropylamine (300 μL; 2.0 mmol). Yield: 87 mg (-30%).
[0289] LCMS m/z 425 [M+H]+, 427 [M+2+H]+ , HPLC tR = 5.40 min
Example 73: l l-Chloro-Σ-fluoro-dibenzofάJiri^jthiazepine-δ-carboxylic acid (3- chlorobenzyD-amide
Figure imgf000087_0002
[0290] The title compound was synthesized by the general procedure for amide formation using l l-chloro-4-fluoro-dibenzo[6,/][l,4]thiazepine-8-carbonyl chloride (~ 0.35 mmol), 8 mL dry dichloromethane and 3-chlorobenzylamine (252 μL; 2.0 mmol). Yield: 117 mg (-34%). LCMS m/z 431 [M+H]+, 433 [M+2+H]+ , 436 [M+4+H]+ . HPLC tR = 5.40 min.
Example 74: ll-(4-Chlorophenyl)-2-fluoro-dibenzo[^,/][l,41thiazepine-8-carboxylic acid Ω-phenylpropylVamide
Figure imgf000088_0001
[0291] The title compound was synthesized by the general procedure for palladium catalyzed Negishi cross-coupling of amidoimidoyl chlorides and arylzinc halides using ll-chloro-2-fluoro-dibenzo[δ,/][l,4]thiazepine-8-carboxylic acid (2-phenylpropyl)- amide (29 mg; 0.067 mmol) and 4-chlorophenylzinc iodide (0.5 M in THF). The compound was purified by preparative HPLC. Yield: 4.3 mg. LCMS m/z 501 [M+H]+, 503 [M+2+H]+. HPLC tR = 6.68 min.
Example 75: 1 l-(3-Chlorophenyl")-2-fluoro-dibenzo[6,/1[l,4]thiazepine-8-carboxylic acid (2-phenylpropylVamide
Figure imgf000088_0002
[0292] The title compound was synthesised by the general procedure for palladium catalyzed Negishi cross-coupling of amidoimidoyl chlorides and arylzinc halides using ll-chloro-2-fluoro-dibenzo[έj][l,4]thiazepine-8-carboxylic acid (2-phenylpropyl)- amide (29 mg; 0.067 mmol) and 3-chlorophenylzinc bromide (0.5 M in THF). The compound was purified by preparative HPLC. Yield: 5.3 mg. LCMS m/z 501 [M+H]+, 503 [M+2+H]+. HPLC fa = 6.65 min.
Example 76: 2-Fluoro-l l-piperidin-l-yl-dibenzo|"6,/][l,4]thiazepine-8-carboxylic acid (2- phenylpropyD-amide
Figure imgf000089_0001
[0293] The title compound was synthesised by the general procedure for synthesis of amidines using l l-chloro-2-fluoro-dibenzo[6,/][l,4]thiazepine-8-carboxylic acid (2- phenylpropyl)-amide (29 mg; 0.067 mmol) and piperidine. The title compound was purified by preparative HPLC. Yield: 8.3 mg. LCMS m/z 474 [M+H]+. HPLC tR = 5.65 min.
Example 77: ll-(3-Chlorophenyl)-2-fluoro-dibenzo|"6,/]ri,41thiazepine-8-carboxylic acid (3 -chlorobenzvD-amide
Figure imgf000089_0002
[0294] The title compound was synthesised by the general procedure for palladium catalyzed Negishi cross-coupling of amidoimidoyl chlorides and arylzinc halides using l lκ;hloro-2-fluoro-dibenzo[&,/J[l,4]thiazepine-8-carboxyric acid (3-chlorobenzyl)- amide (22 mg; 0.052 mmol) and 3-chlorophenylzinc bromide (0.5 M in THF). The compound was purified by preparative HPLC. Yield: 2.3 mg. LCMS m/z 507 [M+H]+, 509 [M+2+H]+. HPLC tR = 6.73 min.
Example 78 : 11 -(4-Chlorophenyl)-2-fluoro-dibenzo|"fr, f] [1 ,41thiazepine-8-carboxylic acid (3 -chlorobenzyD-amide
Figure imgf000090_0001
[0295] The title compound was synthesised by the general procedure for palladium catalyzed Negishi cross-coupling of amidoimidoyl chlorides and arylzinc halides using l l-chloro-2-fluoro-dibenzo[έ,/J[l,4]thiazepine-8-carboxylic acid (3-chloroberi2yl)- amide (22 mg; 0.052 mmol) and 4-chlorophenylzinc iodide (0.5 M in THF). The compound was purified by preparative HPLC. Yield: 5.6 mg. LCMS m/z 507 [M+H]+, 509 [M+2+H]+. , HPLC /R = 6.78 min.
Example 79: l l-Cyclohexyl-2-fluoro-dibenzo[6,/][l,4]thiazepine-8-carboxylic acid (3- chlorobenzvD-amide
Figure imgf000090_0002
[0296] The title compound was synthesised by the general procedure for iron catalyzed cross-couplings using ll-chloro-2-fluoro-dibenzo[δJ/J[l,4]thiazepine-8-carboxylic acid (3-chlorobenzyl)-amide (22 mg; 0.052 mmol) and cyclohexylmagnesium chloride (2 M in diethyl ether). The compound was purified by preparative HPLC. Yield: 5.7 mg. LCMS m/z 479 [M+H]+, 481 [M+2+H]+. HPLC tR = 7.37 min. Example 80: 2-Fluoro-l l-piperidin-l-yl-dibenzo[έ>,/iri,4]thiazepine-8-carboxylic acid (3- chlorobenzvD-amide
Figure imgf000091_0001
[0297] The title compound was synthesised by the general procedure for synthesis of amidines using l l-chloro-2-fluoro-dibenzo[δ,/J[l,4]thiazepine-8-carboxylic acid (3- chlorobenzyl)-amide (22 mg; 0.052 mmol) and piperidine. The title compound was purified by preparative HPLC. Yield: 6.5 mg. LCMS m/z 480 [M+H]+. HPLC tκ = 5.77 min.
Synthesis of 3-fluoro and 3-chloro analogs
Examples 81-104
[0298] The synthesis of 3-fluoro and 3-chloro analogs are synthesized using 4- fluoro-2-mercaptobenzoic acid and 3-chloro-2-mercaptobenzoic acid according to the procedures in Marciano et al., Bioorg, Med. Chem. Lett. (1997), 7, 1709-1714, which is incorporated by reference in its entirety.
[0299] The following compounds are examples several of 3-fluoro and 3-chloro analogs:
Figure imgf000092_0001
Figure imgf000092_0002
Figure imgf000092_0003
Figure imgf000093_0001
Figure imgf000093_0002
Figure imgf000093_0003
Alternative synthesis of 3-chloro analogs:
Scheme 8:
Figure imgf000094_0001
Example 105: 4-fer^Butylsulfanyl-3-nitrobenzoic acid ethyl ester
Figure imgf000094_0002
[0300] As shown in Scheme 8, ethyl 4-fluoro-3-nitrobenzoate (3.86 g; 18.1 mmol) was dissolved in 90 mL dry DMF together with cesium carbonate (11.8 g; 36.2 mmol). tørt-Butylmercaptane (8.15 mL; 72.4 mmol) was added and the mixture was stirred at room temperature for 45 min. Water (50 mL) and ethyl acetate (50 mL) was added and the phases separated. The organic layer was washed with water (2 x 50 mL) followed by drying over magnesium sulfate. After filtration and evaporation 4.95 g (97 %) of a crude yellow oil was isolated that was used without further purification.
[0301] Rf = 0.25 (EtOAc/heptane 30:70). 1H NMR (CDCl3, 400 MHz) δ 8.32 (d, IH, J= 2.0, ArH6), 8.10 (dd, IH, J= 2.0, 8.0, ArH2), 7.75 (d, IH, J= 8.0, ArH5), 4.37 (q, 2H, J= 7.2, OCH2), 1.38 (t, 3H, J= 7.2, CH3), 1.35 (s, 9H, /Bu). Example 106: 4-Mercapto-3 -nitrobenzene acid ethyl ester
Figure imgf000095_0001
[0302] Trifluoroacetic acid (90 rnL) was added to a solution of 4-tert- butylsulfanyl-3-nitrobenzoic acid ethyl ester (4.65 g; 16.4 mmol) in 20 mL dichloromethane. The mixture was stirred for 3 days at room temperature before evaporation of the solvent.
The residue was partitioned between dichloromethane and IM aqueous sodium carbonate.
< After acidification of the aqueous phase using 4M HCl the desired compound was extracted from the aqueous layer with ethyl acetate. The organic layer was dried over sodium sulfate, filtered and evaporated to dryness. The crude compound was used in the next step without purification (1.86 g, 50%).
Example 107: 2-(4-(Ethoxycarbonyl)-2-nitrophenylsulfanyl)-4-chlorobenzoic acid
Figure imgf000095_0002
[0303] To a mixture of 4-chloro-2-iodobenzoic acid (1.02 g; 3.62 mmol), copper(I)iodide (72.2 mg; 0.17 mmol) and potassium carbonate (947 mg; 6.82 mmol) under argon was added 4-mercapto-3-nitrobenzoic acid ethyl ester (776 mg; 3.41 mmol), ethylene glycol (380 μL; 6.82 mmol) and 10 mL 2-propanol. The mixture was stirred at 800C for VAh before cooling to room temperature where stirring was attained overnight. Water, 4M HCl and ethyl acetate were added. After separation of the phases the organic phase was washed several times with water, before drying over magnesium sulfate and concentration in vacuo. Purification was done by silica gel column chromatography (0-8% methanol in dichloromethane) to afford the desired compound as yellow crystals (921 mg; 71%).
[0304] 1H NMR (CDCl3, 400 MHz) δ 8.80 (d, IH, J= 2.0, ArH), 8.10 - 8.02 (m, 2H5 ArH), 7.54 - 7.51 (m, 2H, ArH), 7.07 (d, IH, J = 8.8, ArH), 4.41 (q, 2H, J = 7.2, OCH2), 1.41 (t, 3H, J = 7.2, CH3). LCMS m/z 399 [M+NH4]+, purity (UV/MS) 94/84, fc= 7.86 miη.
Example 108: 4-(3-Chloro-6-carboxyphenylsulfanylV3-nitrobenzoic acid
Figure imgf000096_0001
[0305] 2-(4-(Ethoxycarbonyl)-2-nitrophenylsulfanyl)-4-chlorobenzoic acid (892 mg; 2.34 mmol) was dissolved in a mixture of IM LiOH (aq, 11 mL) and THF (35 mL). The reaction mixture was stirred at 700C for 4 hours. Upon addition of 4M HCl a yellow oil precipitated from the aqueous layer which was extracted with ethyl acetate. The organic layer was dried over magnesium sulfate, filtered and evaporated to dryness affording 1.25 g of which only the majority could be dissolved in ethyl acetate leaving a white solid. After filtration precipitates were accomplished with copious amounts of heptane to afford the title compound as a yellow solid (682 mg; 82%).
[0306] LCMS m/z 371 [M+NH4]+, tR = 0.67 min.
Example 109: 3-Amino-4-(3-chloro-6-carboxyphenylsulfanyl)benzoic acid
Figure imgf000096_0002
[0307] A solution of 4-(3-chloro-6-carboxyphenylsulfanyl)-3-nitrobenzoic acid (680 mg; 1.92mmol) and potassium carbonate (1.32 g; 9.61 mmol) in 40 mL water was cooled to O0C. Sodium dithionite (1.67 g; 9.61 mmol) was added portionwise over 5 min. When the shiny yellow colour had disappeared the reaction mixture was allowed to reach room temperature. Drops of 4M HCl were added until precipitates appeared. Ethyl acetate was added (10 mL) and after separation of the layers the organic phase was concentrated in vacuo to afford the title compound as a white crystalline solid. Used immediately without purification. Example 110: 3-Chloro-l l-oxo-10,ll-dihvdro-dibenzo[&,/1fL41thiazepine-8-carboxylic acid
Figure imgf000097_0001
[0308] 3-Amino-4-(3-chloro-6-carboxyphenylsulfanyl)benzoic acid (-1.92 mmol) was dissolved in 20 mLdry THF at room temperature. 1,1-Carbonyldiimidazole (1.51 g; 1.52 mmol) was added portionwise and the mixture stirred at room temperature for 2 hours. 4 mL 4M HCl was added ensued by 10 mL of water. The colourless precipitate was collected by filtration to afford the desired compound as a white solid (159 mg; 27% over two steps).
[0309] LCMS m/z 306 [M+H]+, purity (UV/MS) 98/-, tR = 3.47 min.
Example 111: 1 l-Chloro-3-chloro-dibenzo[&,/][l,41thiazepine-8-carboxylic acid butyl amide
Figure imgf000097_0002
[0310] 3-Chloro-l l-oxo-10,1 l-dihydro-dibenzo[ό,/][l,4]thiazepine-8-carboxylic acid (38.5 mg; 0.13 mmol), thionyl chloride (2 mL), JV,N-dimethylformamide (100 μL) and toluene (2 mL) was heated to 1000C for 4 hours. The crude mixture was concentrated to dryness to leave the crude acid and imidoyl chloride. The trichloride was redissolved in 5 mL dry dichloromethane and cooled to 00C. A solution of π-butyl amine (37 μL; 0.38 mmol) in 2 mL dry dichloromethane was added and the mixture stirred for 1 hour. After evaporation of the solvent the residue was purified by silica gel column chromatography (0-30% ethyl acetate in heptane) to afford 27.5 mg of a white solid (58 %).
[0311] LCMS m/z 379 [M+H]+, purity (UV/MS) 100/100, tR= 4.70 min.
Example 112: l l-(4-Chlorophenyl)-3-chloro-dibenzo|"6,/][l,41thiazepine-8-carboxylic acid butyl amide
Figure imgf000098_0001
[0312] A reaction flask was charged with l l-Chloro-3-chloro-dibenzo[έ,/j [l,4]thiazepine-8-carboxylic acid butyl amide (27.5 mg; 0.073 mmol) and bis(triphenylphosphine) palladium(II) chloride (3.3 mg; 0.047 mmol) under argon. 4 mL dry tetrahydrofuran was added and followed by addition of 4-chlorophenylzinc iodide (0.5 M in tetrahydrofuran, 290 μL; 0.145 mmol) at room temperature. The mixture was stirred for 1A hour before evaporation of the solvent. The crude residue was purified by silica gel column chromatography (0-10% ethyl acetate in heptane) to afford the title compound as a yellow oil (25.7 mg; 78 %).
[0313] 1H NMR (CDCl3, 400 MHz) δ 7.75 - 7.71 (m, 2H, ArH), 7.66 (t, IH, J = 1.2, ArH), 7.57 (d, IH, J= 2.0, ArH), 7.51 (d, 2H, J= 1.2, ArH), 7.44 - 7.40 (m, 2H, ArH), 7.30 (dd, IH, J= 2.0, 8.4, ArH), 7.10 (d, IH, J= 8.0, ArH), 6.09 (br m, IH, NH), 3.47 - 3.41 (m, 2H, NCH2), 1.63 - 1.54 (m, 2H, CH2), 1.46 - 1.35 (m, 2H, CH2), 0.95 (t, 3H, J= 7.2, CH3). 13C NMR (CDCl3, 100 MHz) δ 167.3, 166.6, 148.8, 141.9, 138.3, 137.7, 136.4, 135.3, 133.0, 132.3, 131.5, 131.4, 131.1*, 128.9*, 128.8, 124.6, 124.0, 40.1, 31.9, 20.3, 14.0. *Denotes double intensity. LCMS m/z 454 [M+H]+, purity (UV/MS) 100/77, tR= 6.88 min.
Synthesis of sulfoxide and sulfone analogs:
[0314] The sulfoxides and sulfones described below (Examples 113 - 121) were synthesized from compounds that have been described previously.
Example 113: N-( 4-FluorobenzvD- l l-C4-chlorophenyr)-5-oxo-5H-5λ4- dibenzo \b,f\\\ .41thiazepine-8-carboxamide
Figure imgf000099_0001
[0315] N-(4-Fluoroben2yl)-l l-(4-chlorophenyl)-dibenzo[&,/][l,4]thiazepine-8- carboxamide (182 mg, 0.385 mmol) was suspended in acetic acid (25 mL). Hydrogen peroxide (35% aqueous solution: 1.65 mL) was added dropwise to the suspension at room temperature. After ~ 5 hours stirring at room temperature the reaction mixture became clear yellow solution. The stirring was continued overnight at room temperature. The reaction mixture was slowly poured into saturated aqueous sodium bicarbonate (150 mL) - vigorous gas liberation. The neutralized mixture (pH~7) was extracted with DCM. The organic layer was washed with saturated aqueous sodium bicarbonate, dried over sodium sulphate, filtered and evaporated to dryness. The residue was a mixture of the desired product and the corresponding 5,5-dioxo compound. Purification of the crude mixture by silica gel column chromatography, eluting with a stepwise gradient of 10-30% ethyl acetate in toluene, afforded the desired compound (54 mg, 29 %). R/= 0.20 (EtOAc/Toluene 20:80).
[0316] 1H ΝMR (CDCl3, 300 MHz) δ 7.91-7.70 (m, 7H, Ar-H), 7.50-7.44 (m, 3H, Ar-H), 7.35-7.27 (m, 3H, Ar-H), 7.03 (m, 2H, Ar-H), 6.56 (m, IH, NH), 4.61 (m, 2H, CH2PhF). LCMS m/z 489 [M+H]+ HPLC ^= 5.1 min.
Example 114: N-f4-FluorobenzylV 1 l-f4-chlorophenyiy5,5-dioxo-5H-5λ6- dibenzo [6,/1[1 ,4]thiazepine-8-carboxamide
Figure imgf000099_0002
[0317] The desired compound was isolated from the crude mixture, which was obtained during the preparation of N-(4-fluorobenzyl)-l l-(4-chlorophenyl)-5-oxo-5H-5λ4- dibenzo[fr/][l,4]thiazepine-8-carboxamide. Purification by silica gel column chromatography eluting with a stepwise gradient of 10-30% ethyl acetate in toluene, afforded the desired compound (46 mg, 23 %). R/= 0.41 (EtOAc/Toluene 20:80).
[0318] 1H NMR (CDCl3, 300 MHz) δ 8.19-8.15 (m, 2H, Ar-H), 7.89-7.66 (m, 6H, Ar-H), 7.47 (m, 3H, Ar-H), 7.33 (m, 2H, Ar-H), 7.06 (m, 2H, Ar-H), 6.50 (m, IH, NH), 4.63 (m, 2H5 CH2PhF). LCMS m/z 505 [M+H]+ HPLC rR= 5.1 min.
Example 115: N-rS-ChlorobenzvD-l l-^-fluorophenylVS-oxo-SH-Sλ4- dibenzofά, f\\\ ,41thiazepine-8-carboxamide
Figure imgf000100_0001
[0319] N-(3-Chlorobenzyl)-l l-(4-fluorophenyl)-dibenzo[6,/][l,4]thiazepine-8- carboxamide (25 mg; 0.05 mmol) was dissolved in DCM (3 mL) and 3-chloroperbenzoic acid (26 mg; 0.15 mmol) was added. The mixture was stirred at room temperature for 1 hour. At this point TLC showed full conversion of the starting material and formation of 2 products. The reaction mixture was diluted with DCM and washed three times with saturated aqueous sodium bicarbonate to extract excess 3-chloroperbenzoic acid. The organic phase was dried over sodium sulphate, filtered and evaporated to dryness. Purification was done by silica gel column chromatography eluting with 20-50 % ethyl acetate in heptane to give the title compound (9.9 mg).
[0320] 1H NMR (acetone-^, 400 MHz) δ 8.42 (br s, IH), 8.01-7.95 (m, 3H), 7.90-7.83 (m, 3H), 7.75 (d, IH, J= 8.0), 7.61 (m, IH), 7.44-7.40 (m, 2H), 7.34-7.25 (m, 4H), 4.61 (d, 2H, J= 6.0). LCMS m/z 489 [M+H]+, 491 [M+2+H]+ HPLC tR = 4.97 min.
Example 116: 3-Chlorobenzyl)-l l-(4-fluorophenylV5.5-dioxo-5H-5λ6- dibenzor&,/iπ,41thiazepine-8-carboxamide
Figure imgf000101_0001
[0321] The desired compound was isolated from the crude mixture, which was obtained during the preparation of N-(3-chlorobenzyl)-ll-(4-fluorophenyl)-5-oxo-5H-5λ4- dibenzo[όj][l,4]thiazepine-8-carboxarnide. Purification by silica gel column chromatography eluting with a stepwise gradient of 20-50% ethyl acetate in heptane, afforded the desired compound (2.3 mg).
[0322] 1H ΝMR (acetone-^, 400 MHz) δ 8.18-8.06 (m, 3H), 7.98-7.85 (m, 5H), 7.65-7.62 (m, IH), 7.44-7.42 (m, IH), 7.36-7.26 (m, 5H), 4.66-4.61 (m, 2H), 3.44 (q, 2H, J= 7.2), 1.58 (m, 2H, J = 7.2), 1.39 (m, 2H, J = 1.2), 0.94 (t, 3H, J= 7.2). LCMS m/z 505 [M+H]+, 507 [M+2+H]+ HPLC tR= 5.08 min.
Example 117: N-butyl-1 l-(4-chlorophenyl)-5-oxo-5H-5λ4-dibenzor6,/iπ.41thiazepine-8- carboxamide
Figure imgf000101_0002
[0323] //-Butyl- 11 -(4-chlorophenyl)-dibenzo[δ,/] [1 ,4]thiazepine-8-carboxamide (86 mg; 0.2 mmol) was dissolved in acetic acid (20 mL) and methanol (15 mL). Hydrogen peroxide (-35% in water; 1 mL) was added. The reaction mixture was stirred at room temperature for 5 hours before it was neutralized by addition of saturated aqueous sodium bicarbonate. The aqueous solution was extracted with DCM (3 x 10 mL) and the combined organic phases were washed with water before drying over sodium sulphate, filtration and evaporation of the solvent in vacuo. The resulting residue was purified by silica gel column chromatography (20 - 50% ethyl acetate in heptane) followed by preparative TLC on silica eluting 4 times with 5% ethyl acetate in heptane to give the desired compound (20.1 mg; 23%). [0324] 1H NMR (CDCl3, 400 MHz) δ 7.92 - 7.89 (m, IH), 7.80-7.76 (m, 3H), 7.74-7.68 (m, 3H), 7.49 - 7.42 (m, 3H), 7.26 (dd, IH, J= 0.8, 7.6), 6.21 (m, IH), 3.44 (q, 2H, J= 7.2), 1.58 (m, 2H, J = 7.2), 1.39 (m, 2H, J= 7.2), 0.94 (t, 3H, J= 7.2). LCMS m/z A31 [M+H]+, 439 [M+2+H]+ HPLC tR= 4.83 min.
Example 118: N-butyl-1 l-r4-chlorophenyl)-5.5-dioxo-5H-5λ6-dibenzor5,/l[1.41thiazepine-8- carboxamide
Figure imgf000102_0001
[0325] N-Butyl-11 -(4-chlorophenyl)-dibenzo[fr/J [1 ,4]thiazepine-8-carboxamide (70 mg; 0.17 mmol) was dissolved in DCM (10 niL) and 3-chloroperbenzoic acid (225 mg; 1.0 mmol) was added. After 4 hours stirring at room temperature the mixture was diluted with DCM (20 mL) and washed with saturated aqueous sodium hydrogen carbonate (3 x 15 mL). The organic phase was dried over sodium sulphate, filtered and evaporated to dryness. Purification by preparative TLC eluting twice with 50% ethyl acetate in heptane afforded the title compound (7.9 mg; 10 %).
[0326] 1H ΝMR (acetone-^, 400 MHz) δ 8.18 - 8.13 (m, IH), 8.08 (d, IH, J = 8.0), 8.01 (d, IH, J= 1.6), 7.94 - 7.86 (m, 5H), 7.67m - 7.58 (m, 3H), 3.42 (q, 2H, J = 7.4), 1.60 (qn, 2H, J= 7.4), 1.40 (m, 2H, J= 7.4), 0.93 (t, 3H, J= 7.4). LCMS m/z 453 [M+H]+, 455 [M+2+H]+ HPLC tR = 7.93 min.
Example 119: l l-(l-Oxy-piperidin-l-yl)-dibenzo[6,/]ri,41thiazepine-8-carboxylic acid 3- chlorobenzylamide (A) and 5-oxo-l l-piperidin-l-yl-5H-5λ4-dibenzo[6,/]-|"l,41thiazepine-8- carboxylic acid 3-chlorobenzylamide (B)
Figure imgf000102_0002
[0327] l l-Piperidinyl-dibenzo[δ,/J[l,4]thiazepine-8-carboxylic acid 3- chlorobenzyl-amide (280 mg; 0.61 mmol) was dissolved in acetic acid (20 mL) and hydrogen peroxide (~35% in water; 2 mL) added. The mixture was stirred at room temperature for 5 hours. The reaction mixture was neutralized by addition of aqueous saturated NaHCO3. The aqueous solution was extracted with DCM (3 x 10 mL) and the combined organic phases were washed with water before drying over sodium sulphate, filtration and evaporation of the solvent in vacuo. Formation of two products was observed by TLC (A: R/ 0.06; B: R/ 0.25; 1:1 EtO Ac/heptane). Both products were isolated by preparative TLC on aluminium oxide eluting twice with 50% ethyl acetate in heptane. Yield: A: 3.0 mg; B: 33 mg as a fine white powder.
[0328] A: LCMS m/z 478 [M+H]+, 480 [M+2+H]+. HPLC fe = 4.13 min.
[0329] B: 1H NMR (400 MHz, CD3Cl) δ 7.83 (dd, IH, J= 1.2, 7.6), 7.63 - 7.57 (m, 2H), 7.53 (dd, IH, J= 2.0, 8.4), 7.44 (dt, IH, J= 1.2, 7.6), 7.39 (d, IH, J= 1.6), 7.31 (dd, IH, J= 1.2, 7.6), 7.29 - 7.15 (m, 4H), 6.64 (m, IH), 4.55 (d, 2H, J= 6.0), 3.85 - 3.30 (br s, 2H), 1.72 - 1.45 (m, 8H). LCMS m/z 478 [M+H]+, 480 [M+2+H]+. HPLC tκ = 4.65 min.
Example 120: 5,5-Dioxo-l l-piperidin-l-yl-5H-5λ4-dibenzo[6,/1[l,4]thiazepine-8-carboxylic acid 3-chlorobenzylamide
Figure imgf000103_0001
[0330] l l-Piperidinyl-dibenzo[έj][l,4]thiazepine-8-carboxylic acid 3- chlorobenzyl-amide (259 mg; 0.56 mmol) was dissolved in DCM (15 mL) and 3- chloroperbenzoic acid (275 mg; 1.23 mmol) was added. The mixture was stirred at room temperature for 3 hours. The mixture was diluted with 20 mL DCM and washed with saturated aqueous NaHCO3 (3 x 15 mL) before drying over sodium sulphate, filtration and removal of the solvent by evaporation under reduced pressure. The crude product was purified by preparative TLC on silica eluting twice with 10% ethyl acetate in heptane to give the title compound (33 mg; 12%). [0331] 1H NMR (400 MHz, CD3Cl) δ 8.00 (d, IH, J = 8.0), 7.92 (d, IH, J = 8.4), 7.64 (m, 2H), 7.52-7.45 (m, 2H), 7.41 (m, IH), 7.30-7.17 (m, 4H), 6.46 (m, IH), 4.60-4.55 (m, 2H), 3.49 (br s, 2H), 1.92-1.44 (m, 8H). LCMS m/z 494 [M+H]+, 496 [M+2+H]+. HPLC fo = 4.93 min.
Example 121: 1 l-Cvclohexyl-5,5-dioxo-5H-5λ4-dibenzo[6,/]["l,41thiazepine-8-carboxylic acid 4-fluorobenzylamide
Figure imgf000104_0001
[0332] l l-Cyclohexyl-dibenzo[6,/]-[l,4]thiazepine-8-carboxylic acid (4- fluorobenzyl)amide (110 mg; 0.25 mmol) was dissolved in DCM (10 mL) and 3- chloroperbenzoic acid (84 mg; 0.37 mmol) was added. The mixture was stirred at room temperature for 2 hours. The mixture was diluted with 10 mL DCM and washed with saturated aqueous NaHCO3 (3 x 10 mL) before drying over sodium sulphate, filtration and removal of the solvent by evaporation under reduced pressure. The crude product was purified by preparative TLC on silica eluting 4 times with 5% EtOAc in heptane to give the title compound (2.2 mg). LCMS m/z All [M+H]+. HPLC fR = 5.25 min.
Synthesis of Nitrogen Analogs:
Example 122: 8-Chlorό-l l-(4-fluorophenyl)-5H-dibenzor&,e1[T,41diazepine
Figure imgf000104_0002
[0333] Bis(triphenylphosphine)palladium(II) chloride was added to a solution of 8,l l-dichloro-5H-dibenzo[δ,e][l,4]diazepine (100 mg, 0.38 mmol) in anhydrous TΗF (10 mL) at room temperature under argon atmosphere, followed by addition of 4- fluorophenylzinc bromide (2.28 ml, 1.14 mmol). After 3 hours stirring at room temperature the reaction mixture was partitioned between saturated aqueous ammonium chloride and ethyl acetate. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 30% ethyl acetate in n-heptane, afforded the desired product (88 mg, 72%). R/= 0.38 (EtO AcIn- Heptane 30:70). LCMS m/z 323 [M+H]+ HPLC tκ= 5.6 min.
Example 123: N-(4-FluorobenzylVl l-(4-fluorophenyl)-5H-dibenzo[ά,e]f L4]diazepine-8- carboxamide
Figure imgf000105_0001
[0334] The desired compound was synthesized using a literature procedure in Lagerlund et al., J Comb. Chem. (2006), 8, 4-6, which is hereby incorporated by reference in its entirety. 8-Chloro-ll-(4-fluorophenyl)-5H-dibenzo[δ,e][l,4]diazepine (40 mg, 0.12 mmol) was reacted with 4-fluorobenzylamine (46 mg, 0.37 mmol), molybdenum hexacarbonyl (32 mg, 0.12 mmol), trans-di-(μ-acetato)-bis[o-(di-o-tolylphosphino) benzyl]dipalladium(II) (2.3 mg, 0.025 mmol), tri-tert-butylphosphine tetrafluoroborate (1.7 mg, 0.05 mmol), and l,8-diazabicyclo[5.4.0]undec-7-ene (56 mg, 0.37 mmol) in anhydrous TΗF (0.5 mL). The reaction mixture was heated in a sealed flask for 20 minutes at 170° C under microwave irradiation. The reaction mixture was partitioned between DCM and weak acidic aqueous layer (10 mL water was acidified with 2-3 drops of concentrated HCl). The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue using a silica gel column chromatography, eluting with a stepwise gradient of 20 to 50% ethyl acetate in «-heptane, afforded the title compound (16 mg, 30%). Ry = 0.19 (EtOAc/n-Ηeptane 50:50).
[0335] 1H NMR (CDCl3, 300 MHz) δ 7.75-7.53 (m, 4H, Ar-H), 7.40-7.26 (m, 3H, Ar-H), 7.18-6.92 (m, 6H, Ar-H), 6.87-6.76 (m, 2H, Ar-H), 6.69-6.54 (m, IH, NH), 5.79-5.56 (m, IH, NH), 4.59 (m, 2H, CH2PhF). LCMS m/z 440 [M+H]+. HPLC tR= 4.6 min. Example 124: N-Butyl-1 l-r4-fluorophenylV5H-dibenzo["ά,e][1.41diazepine-8-carboxamide
Figure imgf000106_0001
[0336] The title compound was synthesized from 8-chloro-l l-(4-fluorophenyl)- 5H-dibenzo[ό,e][l,4]diazepine (25 mg, 0.077 mmol) and rø-butylamine (17 mg, 0.23 mmol) using the same procedure as for synthesis of N-(4-fluorobenzy I)-I l -(4-fluoropheny 1)-5H- dibenzo[b,e][l,4]diazepine-8-carboxamide. R/= 0.32 (EtOAc/«-Ηeptane 50:50). LCMS m/z 388 [MH-H]+.. HPLC /R = 4.4 min.
Example 125: 1 l-(4-Fluorophenyl)-N-(l-phenylethylV5H-dibenzo|'&,gl[l,41diazepme-8- carboxamide
Figure imgf000106_0002
[0337] The title compound was synthesized from 8-chloro-l 1 -(4-fluoropheny I)- 5H-dibenzo[δ,e][l,4]diazepine (25 mg, 0.077 mmol) and DL-1-phenylethyl amine (28 mg, 0.23 mmol) using the same procedure as for synthesis of JV-(4-fluorobenzyl)-l l-(4- fluorophenyl)-5H-dibenzo[δ,e][l,4]diazepine-8-carboxamide. R/ = 0.33 (EtOAc/n-Ηeptane 50:50). LCMS m/z 436 [M+Η]+, HPLC fe= 4.7 min.
Example 126: 8-Chloro-l l-(4-fluorophenyl)-5-methyl-5H-dibenzo[6,gl[l,41diazepine
Figure imgf000106_0003
[0338] Sodium hydride (60% suspension in an mineral oil: 18 mg, 0.38 mmol) was added to a solution of 8-chloro-l l-(4-fluorophenyl)-5H-dibenzo[6,e][l,4]diazepine (60 mg, 0.19 mmol) in dry DMF (2 mL) at room temperature. After 10 minutes shaking at room temperature, the reaction mixture became green and iodomethane (25 μX, 0.38 mmol) was added. The reaction mixture was shaken for 2 hours at 500C and then at room temperature overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with 4% aqueous magnesium sulphate, dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 10% ethyl acetate in rc-heptane, afforded the title compound (40 mg, 60%). R/= 0.47 (EtOAc/rc-Heptane 30:70). LCMS m/z 337 [M+H]+.. HPLC tκ= 6.4 min.
Example 127: N-f4-Fluorobenzyl)-l l-f4-fluorophenyl)-5-methyl-5H- dibenzo \b, el F 1 ,41 diazepine -8 -carboxamide
Figure imgf000107_0001
[0339] The title compound was synthesized from 8-chloro-l l-(4-fluorophenyl)-5- methyl-5H-dibenzo[6,e][l,4]diazepine (20 mg, 0.060 mmol) and 4-fluorobenzyl amine (22 mg, 0.18 mmol) using the same procedure as for synthesis of N-(4-fluorobenzyl)-l l-(4- fluorophenyl)-5H-dibenzo[δ,e][l,4]diazepine-8-carboxamide. R/ = 0.32 (EtOAc/rø-Ηeptane 50:50). LCMS m/z 454 [M+Η]+.. HPLC tR= 5.0 min.
Example 128: ir8-Chloro-l l-(4-fluorophenyl)-dibenzor6,eiri,41diazepin-5-yllethanone
Figure imgf000107_0002
[0340] N,N-Dimethyl amine (40 mg, 0.33 mmol) was added to a solution of 8- chloro-l l-(4-fluorophenyl)-5H-dibenzo[δ,e][l,4]diazepine (108 mg, 0.33 mmol) in dry TΗF (2 mL) at room temperature, followed by addition of acetyl chloride (70 μL, 0.99 mmol). The reaction mixture was shaken overnight at 600C, allowed to cool to room temperature and partitioned between ethyl acetate and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. The crude mixture was passed over a short silica gel column using a mixture of ethyl acetate and n-heptane (30:70) as the eluant. The isolated fractions were a mixture of the desired compound and a side product. The fractions were left on standing over the weekend. The desired compound was crystallized in the fractions and it was isolated by filtration (69 mg, 60%). R/= 0.20 (EtOAc/n-Heptane 50:50). LCMS m/z 365 [M+H]+.. HPLC rR= 5.0 min.
Examples 129-146
[0341] The following compounds are examples of nitrogen analogs synthesized from 8,l l-dichloro-5H-dibenzo[έ,e][l,4]diazepine according to the general procedure for palladium catalysed Negishi couplings followed by reductive amination and/or alkylation reactions:
Figure imgf000109_0001
Figure imgf000110_0001
Series A
Library synthesis: formation of amidoimidoyl chlorides
[0342] The amidoimidoyl chlorides (Examples 147 - 162) were synthesized according to the general procedure for amide formation at 0.5 mmol scale except that the reaction mixture was passed through a pad of acidic alumina oxide and eluted with a mixture Of CH2Cl2 and EtOAc. The eluents were concentrated at reduced pressure and the obtained crude products were directly used in the next reactions without further purifications or characterization.
Example 147: 1 l-(chloro)-dibenzo[b,f][l,4]thiazepin-8-yl-(piperidin-l-yl)-methanone
Figure imgf000111_0001
[0343] 173 mg
Example 148: N-benzyl-1 l-(chloro)-dibenzo[b,f|[L41thiazepine-8-carboxamide
Figure imgf000111_0002
[0344] 148 mg
Example 149: N-(l-phenylethylVl l-rchloro)-dibenzorb.fiπ.41thiazepine-8-carboxamide
Figure imgf000111_0003
[0345] 168 mg Example 150: N-(butyl)-l l-(chloro)-dibenzorb,f1[l,41thiazepine-8-carboxamide
Figure imgf000112_0001
[0346] 138 mg
Example 151: N-(3-phenylpropyl)-l l-(chloroVdibenzo[b,f][l,4]thiazepine-8-carboxamide
Figure imgf000112_0002
[0347] 167 mg
Example 152: N-(2-phenylethyl)-l l-(chloro)-dibenzorb,f][l,4]thiazepine-8-carboxamide
Figure imgf000112_0003
[0348] 160 mg
Example 153: N-fΣ-chlorobenzylVl l-(chloroVdibenzo[b,f|[l,41thiazepine-8-carboxamide
Figure imgf000112_0004
[0349] 161 mg
Example 154: N-(2.4-dichlorobenzvD-l l-rchloroVdibenzorb.f|[1.41thiazepine-8- carboxamide
Figure imgf000113_0001
[0350] 120 mg
Example 155: N-(2-(4-chlorophenyl)ethyl)-l l-(chloro)-dibenzo[b,f1[l,41thiazepine- carboxamide
Figure imgf000113_0002
[0351] 167 mg
Example 156: N-(2-(3 -chlorophenvPethyD- 11 -(chloro)-dibenzo [b,f| [ 1 ,4~|thiazepine - carboxamide
Figure imgf000113_0003
[0352] 171 mg
Example 157: N-(3-chlorobenzyl)-l l-rchloroVdibenzo[b.flfl,4]thiazepine-8-carboxamide
Figure imgf000113_0004
[0353] 176 mg Example 158: N-(2-bromobenzyl)-l l-(chloro)-dibenzo[b.f][l,4]thiazepine-8-carboxaniide
Figure imgf000114_0001
[0354] 180 mg
Example 159: N-(2 -phenyl-propyl)- 1 l-(chloro)-dibenzorb,fiπ,41thiazepine-8-carboxamide
Figure imgf000114_0002
[0355] 172 mg
Example 160: N-((N-ethyl-N-phenyl)aminoethyl)-l l-rchloro)-dibenzo[b,f||"l,41thiazepine-8- carboxamide
Figure imgf000114_0003
[0356] 168 mg
Example 161: l l-rchloro)-dibenzo[b,f|[l,41thiazepin-8-carboxylic acid morpholin-4-yl amide
Figure imgf000114_0004
[0357] 160 mg Example 162: N-(4-fluorobenzyl)-l l-(chloro)-dibenzo[b.f|f l,4]thiazepine-8-carboxamide
Figure imgf000115_0001
[0358] 120 mg
Series B
[0359] The following compounds were prepared according to the general procedure for the synthesis of amidines starting from the appropriate imidoylchloride (15 mg) and piperidine (excess).
Example 163: 1 l-(piperidinyl)-dibenzorb,firi,41thiazepin-8-yl-(piperidin-l-yl)-methanone
Figure imgf000115_0002
[0360] 2.8 mg, UV/MS purity 100/97
Example 164: N-benzyl-1 l-(piperidinylVdibenzorb,firi,41thiazepine-8-carboxamide
Figure imgf000115_0003
[0361] 15.9 mg, UV/MS purity 100/91 Example 165: N-d-phenylethylH Hpiperidinviydibenzo["b,f||~l,4"|thiazepine-8-carboxamide
Figure imgf000116_0001
[0362] 6.2 mg, UV/MS purity 88/54
Example 166: N-(butyl)-l l-fpiperidinyl)-dibenzorb,firiΛlthiazepine-8-carboxamide
Figure imgf000116_0002
[0363] 15.1 mg, UV/MS purity 98/80
Example 167: N-(3-phenylpropyl)-l l-(piperidinyl)-dibenzo[b,f)[l,4]thiazepine- carboxamide
Figure imgf000116_0003
[0364] 16.7 mg mg, UV/MS purity 100/77
?le 168: N-(2-phenylethyl>- 11 -fpiperidinyl)- dibenzo fb,f| \ 1 ,41thiazepine-8-carboxamide
Figure imgf000116_0004
[0365] 14.5 mg, UV/MS purity 99/76 Example 169: N-(2-chlorobenzyl)-l l-Cpiperidinyl)-dibenzo|[b,firi.41thiazepine-8- carboxamide
Figure imgf000117_0001
[0366] 15.2 nig, UV/MS purity 99/73
Example 170: N-(2,4-dichlorobenzyT)-l l-(piperidinyl)-dibenzo[b,f|f l,41thiazepine-8- carboxamide
Figure imgf000117_0002
[0367] 13.2 mg, UV/MS purity 100/73
Example 171 : N-(2-(4-chlorophenyl)ethvD-l l-(piperidinylVdibenzo[b,fl|"l,41thiazepine-8- carboxamide
[0368] 10.7 mg, UV/MS purity 100/79
Example 172: N-(2-(3-chlorophenyl)ethyl)-l l-(piperidinyl)-dibenzo[b.fl[l,4]thiazepine-8- carboxamide
Figure imgf000118_0001
[0369] 8.4 mg, UV/MS purity 99/67
Example 173: N-(3-chlorobenzylVl l-("piperidinyl)-dibenzo[b,f||"l,41thiazepine- carboxamide
Figure imgf000118_0002
[0370] 12.9 mg, UV/MS purity 98/72
Example 174: N-(2-bromobenzylVl l-(piperidinylVdibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000118_0003
[0371] 16.2 mg, UV/MS purity 100/76
Example 175: N-("2-phenyl-propyl)-l l-fpiperidinylVdibenzorb.f||"L41thiazepine-8- carboxamide
Figure imgf000119_0001
[0372] 14.2 mg, UV/MS purity 100/72
Example 176: N-("(N-ethyl-N-phenyl)aminoethyl>-l l-(piperidinyl)- dibenzorb,firi,41thiazepine-8-carboxamide
Figure imgf000119_0002
[0373] 6.0 mg, UV/MS purity 82/60
Example 177: l l-(piperidinyl)-dibenzo[b,f|["1..4]thiazepin-8-carboxylic acid morpholin-4-yl amide
Figure imgf000119_0003
[0374] 5.9 mg, UV/MS purity 100/78
Example 178: N-(4-fluorobenzyl)-l l-(piperidinyl)-dibenzo[b,f|[L41thiazepine-8- carboxamide
Figure imgf000120_0001
[0375] 14.7 mg, UV/MS purity 95/53
Series C
[0376] The following compounds were prepared according to the general procedure for an iron-catalyzed alkyl-imidoyl chloride cross-coupling starting from the appropriate imidoylchloride (15 mg) and cyclohexylmagnesium chloride (6eq). When the reactions were completed saturated ammonium chloride (1 ml) and EtOAc (2 ml) were added to the reaction mixtures. The organic phases were passed through a short silica column (eluted with EtOAc). After concentration at reduced pressure, the obtained crude products were purified by preparative HPLC.
Example 179: 1 l-fcvclohexyD-dibenzorb^firiΛlthiazepin-δ-yl-fpiperidin-l-vD-methanone
Figure imgf000120_0002
[0377] 0.6 mg, UV/MS purity 90/90
Example 180: N-benzyl-1 l-fcyclohexyD-dibenzorb^FMlthiazepine-δ-carboxamide
Figure imgf000120_0003
[0378] 5.1 mg, UV/MS purity 98/83 Example 181 : N-d-phenylethylVl l-fcyclohexylVdibenzofb.fiπ^lthiazepine-δ-carboxamide
Figure imgf000121_0001
[0379] 2.2 mg, UV/MS purity 98/87
Figure imgf000121_0002
[0380] 4.6 mg, UV/MS purity 98/91
Example 183: N-(2-chlorobenzy I)- 11 -(cyclohexyl Vdibenzo f b,f] |" 1 ,4]thiazepine - carboxamide
Figure imgf000121_0003
[0381] 4.5 mg, UV/MS purity 99/85
Example 184: N-f2,4-dichlorobenzyl)-l l-fcvclohexyl)-dibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000122_0001
[0382] 1.8 mg, UV/MS purity 100/82
Example 185: N-(2-(4-chlorophenyl)ethyl)-l l-(cvclohexyl)-dibenzo[b,f][L41thiazepine-8- carboxamide
Figure imgf000122_0002
[0383] 5.9 mg, UV/MS purity 100/87
Example 186: N-(2-f3-chlorophenyl)ethyl)-l l-(cyclohexyl)-dibenzo[b,f][l,41thiazepine-8- carboxamide
Figure imgf000122_0003
[0384] 6.6 mg, UV/MS purity 99/90
Example 187: N-(3-chlorobenzyl)-l l-fcycloheχyl)-dibenzo|'b,fl|"l,41thiazepine-8- carboxamide
Figure imgf000123_0001
[0385] 4.8 mg, UV/MS purity 99/87
Example 188: N-(2-bromobenzyl)-l l-(cyclohexyl)-dibenzo["b,firi.4~|thiazepme-8- carboxamide
Figure imgf000123_0002
[0386] 0.8 mg, UV/MS purity 100/83
Example 189: N-(2-phenyl-propyl)-l l-(cyclohexyl)-dibenzo[b,f|[l,4]thiazepine-^ carboxamide
Figure imgf000123_0003
[0387] 5.3 mg, UV/MS purity 93/83
Example 190: N-((N-ethyl-N-phenyl)aminoethylVl l-(cyclohexyl)- dibenzorb.f] [1 ,41thiazepine-8-carboxamide
Figure imgf000124_0001
[0388] 3.2 mg, UV/MS purity 98/79
Example 191: l l-(cvclohexyl)-dibenzo[b,f1[L4]thiazepin-8-carboxylic acid morρholin-4-yl amide
Figure imgf000124_0002
[0389] 3.8 mg, UV/MS purity 96/75
Example 192: N-f4-fluorobenzyl)-l l-(cvclohexylVdibenzo|"b.,f|[l,41thiazepine-8- carboxamide
Figure imgf000124_0003
[0390] 3.6 mg, UV/MS purity 98/74
Series D-H
[0391] The following compounds were prepared according to the general procedure for Negishi cross coupling starting from the appropriate imidoylchloride (15 mg) and arylzinc halide (8eq). Ammonium chloride (0.02 ml) was added to the reaction mixtures, which were then passed through a short column (Na2SO4/silica) using EtOAc as eluent. The eluents were concentrated at reduced pressure and the crude products were purified by preparative HPLC or by column chromatography (Heptane-EtOAc 4:1-1 :1).
Series D
[0392] The arylzinc halide used for Examples 193 - 205 was 3-chlorophenylzinc iodide.
Example 193: 1 l-(3-chlorophenyl)-dibenzo[b,f|ri,4]thiazepin-8-yl-(piperidin-l-yl)- methanone
Figure imgf000125_0001
[0393] 9.9 mg, UV/MS purity 100/80
Example 194: N-benzyl-1 l-(3-chlorophenyl)-dibenzorb,firi,41thiazepine-8-carboxamide
Figure imgf000125_0002
[0394] 19.2 mg, UV/MS purity 100/60
Example 195: N-Cl-phenylethylH l-f3-chlorophenyl)-dibenzo[b,f][l,4]thiazepine-8- carboxamide
Figure imgf000125_0003
[0395] 16.7 mg, UV/MS purity 100/85 Example 196: N-fbutyl)-l l-(3-chlorophenvl)-dibenzorb.,firi,41thiazepine-8-carboxamide
Figure imgf000126_0001
[0396] 17.3 mg, UV/MS purity 100/79
Example 197: N-Q-phenylpropyD-l l-(3-chlorophenyl)-dibenzo[b,f|["l,41thiazepine-8- carboxamide
Figure imgf000126_0002
[0397] 9.0 mg mg, UV/MS purity 95/80
Example 198: N-f2-phenylethyl)-l l-(3-chlorophenylVdibenzorb,iiri,41thiazepine-8- carboxamide
Figure imgf000126_0003
[0398] 10.9 mg, UV/MS purity 100/80
Example 199: N-(2-chlorobenzyl)-l l-(3-chlorophenyl)-dibenzo[b,f|[l,41thiazepine-8- carboxamide
Figure imgf000127_0001
[0399] 9.7 mg, UV/MS purity 98/80
Example 200: N-(2-(4-chlorophenyl)ethyl>l l-(3-chlorophenyl)-dibenzo[b,f1[L41thiazepine- 8-carboxamide
Figure imgf000127_0002
[0400] 11.2 mg, UV/MS purity 99/76
Example 201 : N-(3-chlorobenzyl)-l l-(3-chlorophenyl)-dibenzo[b,f|["L4]thiazepine-8- carboxamide
Figure imgf000127_0003
[0401] 12.2 mg, UV/MS purity 95/72
Example 202: N-fΣ-phenyl-propyD-l l-O-chlorophenvD-dibenzorb^ri^lthiazepine-S- carboxamide
Figure imgf000127_0004
[0402] 8.8 mg, UV/MS purity 99/59 Example 203: N-((N-ethyl-N-phenyl)aminoethyl>l l-(3-chlorophenyl)- dibenzo[b,ff[l,4]thiazepine-8-carboxarnide
Figure imgf000128_0001
[0403] 6.3 mg, UV/MS purity 97/80
Example 204: l l-(3-chlorophenyl)-dibenzo[b,f|[l,41thiazepin-8-carboxylic acid morpholin- 4-yl amide
Figure imgf000128_0002
[0404] 11.8 mg, UV/MS purity 97/56
Example 205: N-(4-fluorobenzyl)-l l-(3-chlorophenyl)-dibenzo[b,f|[L41thiazepine- carboxamide
Figure imgf000128_0003
[0405] 8.1 mg, UV/MS purity 100/55
Series E
[0406] The arylzinc halide used for Examples 206 - 217 was 4-fluorophenylzinc iodide. Example 206: 1 l-(4-fluorophenyl')-dibenzo[b,f|[l,41thiazepin-8-yl-fpiperidin-l-yl)- methanone
Figure imgf000129_0001
[0407] 9.9 mg, UV/MS purity 99/62
Example 207: N-benzyl-1 l-f4-fluorophenyl)-dibenzo[b,f|fl,4]thiazepine-8-carboxamide
Figure imgf000129_0002
[0408] 12.2 mg, UV/MS purity 96/41
Example 208: N-(l-phenylethyl>l l-(4-fluorophenyl)-dibenzo[b,f|ri^1thiazepine- carboxamide
Figure imgf000129_0003
[0409] 11.4 mg, UV/MS purity 100/91
Example 209: N-CbutylH l-(4-fluorophenyl)-dibenzo[b,firL41thiazepine-8-carboxamide
Figure imgf000130_0001
[0410] 7.5 mg, UV/MS purity 98/93
Example 210: N-(2-phenylethyl>-l l-f4-fluorophenyl)-dibenzorb,f1[1.41thiazepine-8- carboxamide
Figure imgf000130_0002
[0411] 4.6 mg, UV/MS purity 98/62
Example 211 : N-f2-chlorobenzyl>l l-(4-fluorophenylVdibenzo[b,firi,41thiazepine-8- carboxamide
Figure imgf000130_0003
[0412] 8.4 mg, UV/MS purity 100/52
Example 212: N-(2,4-dichlorobenzyl)-l l-f4-fluorophenyl)-dibenzorb,firL41thiazepine-8- carboxamide
Figure imgf000131_0001
[0413] 4.0 mg, UV/MS purity 96/36
Example 213: N-(2-(4-chlorophenyl)ethy IV 11 -(4-fluoropheny D- dibenzo |"b,f] [ 1 ,4]thiazepine - 8-carboxamide
Figure imgf000131_0002
[0414] 5.6 mg, UV/MS purity 100/65
Example 214: N-(2-(3-chlorophenyl)ethyl)-l l-(4-fluorophenylVdibenzo[b,f|ri,41thiazepine- 8-carboxamide
Figure imgf000131_0003
[0415] 1.4 mg, UV/MS purity 99/56 Example 215: N-(3-chlorobenzyl)-l l-(4-fluorophenyl)-dibenzo[b.,fl[l,4]thiazepine-8- carboxamide
Figure imgf000132_0001
[0416] 5.4 mg, UV/MS purity 99/50
Example 216: N-(2-phenyl-propyl")-l l-f4-fluorophenyl)-dibenzorb,f|[l,4]thiazepine-8- carboxamide
Figure imgf000132_0002
[0417] 1.9 mg, UV/MS purity 85/44
Example 217: N-f(N-ethyl-N-phenvDaminoethylH l-(4-fluorophenyl)- dibenzofb,f| [1 ,4"|thiazepine-8-carboxamide
Figure imgf000132_0003
[0418] 1.3 mg, UV/MS purity 78/45
Series F
[0419] The arylzinc halide used for Examples 218 - 232 was 2-fluorophenylzinc iodide. Example 218: 1 l-(2-fluorophenyl)-dibenzo[b,f|[l,41thiazepin-8-yl-(piperidin-l-ylV methanone
Figure imgf000133_0001
[0420] 12.5 mg, UV/MS purity 99/67
Example 219: N-ben2yl-l l-(2-fluorophenyl)-dibenzo|"b,f|ri,4]thiazepine-8-carboxamide
Figure imgf000133_0002
[0421] 13.7 mg, UV/MS purity 100/100
Example 220: N-(I -phenylethy D-I l-(2-fluorophenyl)-dibenzo[b,f]fl,4]thiazepine- carboxamide
Figure imgf000133_0003
[0422] 10.1 mg, UV/MS purity 100/96
Example 221 : N-(butyl)-! l-(2-fluorophenyl)-dibenzorb,firi,41thiazepine-8-carboxamide
Figure imgf000133_0004
[0423] 12.3 mg, UV/MS purity 100/94
Example 222: N-(3-phenylpropyD-l l-(2-fluorophenyl)-dibenzo[b1f|[l,4]thiazepine-8- carboxamide
Figure imgf000134_0001
[0424] 12.3 mg mg, UV/MS purity 100/100
Example 223: N-f2-phenylethylH l-(2-fluorophenyl)-dibenzorb.f|ri,41thiazepine- carboxamide
Figure imgf000134_0002
[0425] 9.3 mg, UV/MS purity 100/100
Example 224: N-(2-chlorobenzylVl l-(2-fluorophenyl)-dibenzo[b,f|[l,41thiazepine- carboxamide
Figure imgf000134_0003
[0426] 12.7 mg, UV/MS purity 100/89 Example 225: N-(2,4-dichlorobenzylVl l-(2-fluorophenylVdiberizo[b,f1[l,4]thiazepine-8- carboxamide
Figure imgf000135_0001
[0427] 10.6 nig, UV/MS purity 100/84
Example 226: N-(2-(4-chlorophenyl)ethyl)-l l-(2-fluorophenyl)-dibenzo|"b.f]|"1.41tliiazepine- 8-carboxamide
Figure imgf000135_0002
[0428] 8.4 mg, UV/MS purity 100/92
Example 227: N-f2-f3-chlorophenyl)ethylVl l-(2-fluorophenyl Vdibenzorb.fi Fl.41thiazepine- 8-carboxamide
Figure imgf000135_0003
[0429] 10.4 mg, UV/MS purity 100/91
Example 228: N-f3-chlorobenzyl)-l l-(2-fluorophenyl)-dibenzorb,f|[l,41thiazepine-8- carboxamide
Figure imgf000136_0001
[0430] 12.5 mg, UV/MS purity 100/95
Example 229: N-(2-bromobenzyl)-l l-f2-fluoroph.enyl)-dibenzo|"b,f|[l,41thiazepine-8- carboxamide
Figure imgf000136_0002
[0431] 8.3 mg, UV/MS purity 100/96
Example 230: N-(2-phenyl-propyl)-l l-(2-fluorophenyl)-dibenzo[b,fl[l,4]thiazepine-8- carboxamide
Figure imgf000136_0003
[0432] 11.2 mg, UV/MS purity 100/90
Example 231 : N-fCN-ethyl-N-phenyPaminoethylH H2-fluorophenviy dibenzo[b,fl [1 ,4"|thiazepine-8-carboxamide
Figure imgf000137_0001
[0433] 5.7 mg, UV/MS purity 100/91
Example 232: N-(4-fluorobenzyl)-l l-f2-fluorophenyl)-dibenzo[b,f|[L41thiazepine- carboxamide
Figure imgf000137_0002
[0434] 12.4 mg, UV/MS purity 100/91
Series G
[0435] The arylzinc halide used for Examples 233 - 246 was phenylzinc iodide.
Example 233: N-benzyl-1 l-fphenyl)-dibenzorb,flri,41thiazepine-8-carboxamide
Figure imgf000137_0003
[0436] 10.2 mg, UV/MS purity 100/57
Example 234: N-d-phenylethyD-1 l-fphenyl)-dibenzo[b,f][l,41thiazepine-8-carboxarnide
Figure imgf000138_0001
[0437] 8.2 mg, UV/MS purity 91/61
Example 235: N-(butyl)-l l-(phenyl)-dibenzorb,f|[l,4]thiazepine-8-carboxaniide
Figure imgf000138_0002
[0438] 9.4 mg, UV/MS purity 94/62
Example 236: N-(3-phenylpropyl)-l l-(phenylVdibenzorb,firL41thiazepine-8-carboxamide
Figure imgf000138_0003
[0439] .. 11.4 mg mg, UV/MS purity 100/100
Example 237: N-(2-phenylethylH l-fphenyl)-dibenzorb,firi,41thiazepine-8-carboxamide
Figure imgf000138_0004
[0440] 9.0 mg, UV/MS purity 97/85 Example 238: N-(2-chlorobenzyl)-l l-Cphenyl)-dibenzo[b,fl[l,41thiazepine-8-carboxamide
Figure imgf000139_0001
[0441] 8.8 mg, UV/MS purity 100/100
Example 239: N-(2,4-dichlorobenzylV 11 -fphenylVdibeozoCb,firi ,41thiazepine-8- carboxamide
Figure imgf000139_0002
[0442] 6.1 mg, UV/MS purity 100/87
Example 240 : N-(2-(4-chlorophenyl)ethyl)- 11 -(phenyl Vdibenzo |"b,f| [ 1 ,41thiazepine - carboxamide
Figure imgf000139_0003
[0443] 9.3 mg, UV/MS purity 100/90
Example 241 : N-(2-(3-chlorophenyl)ethyl)-l l-fphenyl)-dibenzorb.fiπ.41thiazepine- carboxamide
Figure imgf000140_0001
[0444] 8.9 mg, UV/MS purity 100/80
Example 242: N-(3-chlorobenzyl)-l l-(phenyl)-dibenzo|"b,f][l,41thiazepine-8-carboxamide
Figure imgf000140_0002
[0445] 10.1 mg, UV/MS purity 100/100
Example 243: N-f2-bromobenzyl)-l l-(phenyl)-dibenzo[b,f]|"l,4]thiazepine-8-carboxamide
Figure imgf000140_0003
[0446] 10.2 mg, UV/MS purity 100/89
Example 244: N-(2 -phenyl-propyl)- 1 l-(phenyl)-dibenzorb,firi,41thiazeρine-8-carboxamide
Figure imgf000140_0004
[0447] 9.5 mg, UV/MS purity 100/87 Example 245: N-f(N-ethyl-N-phenyl)aminoethylH l-(phenyl)-dibenzo[b,fl[l,4]thiazepine-8- carboxamide
Figure imgf000141_0001
[0448] 10.0 mg, UV/MS purity 100/91
Example 246: N-(4-fluorobenzyl)-l l-(phenyl)-dibenzo|Txf|[M1thiazepine-8-carboxamide
Figure imgf000141_0002
[0449] 12.8 mg, UV/MS purity 100/93
Series H
[0450] The arylzinc halide used for Examples 247 - 260 was 4-chlorophenylzinc iodide.
Example 247: 1 l-(4-chlorophenyl)-dibenzo[b,f|["l,4]thiazepin-8-yl-rpiperidin-l-yl)- methanone
Figure imgf000141_0003
[0451] 2.2 mg, UV/MS purity 100/100 Example 248: N-benzyl-1 l-(4-chlorophenyl)-dibenzo|T3,f|[L41thiazepine-8-carboxamide
Figure imgf000142_0001
[0452] 6.3 mg, UV/MS purity 100/100
Example 249: N-d-phenylethylVl l-(4-chlorophenyl)-dibenzo[b,f][l,4]thiazeρine- carboxamide
Figure imgf000142_0002
[0453] 5.7 mg, UV/MS purity 100/83
Example 250: N-(butyl)-! l-(4-chlorophenyl)-dibenzo[Tλf|[l,4]thiazepine-8-carboxarnide
Figure imgf000142_0003
[0454] 13.7 mg, UV/MS purity 100/100
Example 251 : N-(3-phenylpropyl)-l l-f4-chlorophenyl)-dibenzo[b,fl[l ,4]thiazepine-8- carboxamide
Figure imgf000143_0001
[0455] 12.5 mg, UV/MS purity 100/100
Example 252: N-(2-phenylethyl)-l l-(4-chlorophenyl)-dibenzo[b,f|rh4]thiazepine-8- carboxaniide
Figure imgf000143_0002
[0456] 8.7 mg, UV/MS purity 100/100
Example 253: N-(2-chlorobenzyl)-l l-(4-chlorophenyl)-dibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000143_0003
[0457] 8.4 mg, UV/MS purity 100/100 Example 254: N-(2,4-dichlorobenzyl')-ll-(4-chlorophenyl)-diben2o[b,fH'l,41thiazepine-8- carboxamide
Figure imgf000144_0001
[0458] 5.4 mg, UV/MS purity 100/73
Example 255: N-(2-(4-chlorophenyl)ethyl)-l l-(4-chlorophenyl)-dibenzo[b,f1[l,41thiazepine- 8-carboxamide
Figure imgf000144_0002
[0459] 10.2 mg, UV/MS purity 100/80
Example 256: N-(2-(3-chlorophenyl)ethylVl l-(4-chlorophenyiydibenzoP3,f][L4]thiazepine- 8-carboxamide
Figure imgf000144_0003
[0460] 10.0 mg, UV/MS purity 100/100 Example 257: N-(3-chlorobenzyl)-l l-f4-chlorophenyl)-dibenzo["b,fl[l,41thiazepine-8- carboxamide
Figure imgf000145_0001
[0461] 10.0 mg, UV/MS purity 100/100
Example 258: N-(2-bromobenzyl)-l l-(4-chlorophenyl)-dibenzo[b,f|[l,41thiazepine- carboxamide
Figure imgf000145_0002
[0462] 10.2 mg, UV/MS purity 100/67
Example 259: N-(2-phenyl-propyl)-l l-(4-chlorophenyl')-dibenzo|"b,f|fl,41thiazepine-8- carboxamide
Figure imgf000145_0003
[0463] 11.9 mg, UV/MS purity 100/100
Example 260: N-((N-ethyl-N-phenyl)aminoethyl)-l l-r4-chlorophenyl)- dibenzo[b,f|[l,4]thiazepine-8-carboxamide
Figure imgf000146_0001
[0464] 12.4 mg, UV/MS purity 100/88
Example 261 : N-(4-fluorobenzyl)-l l-(4-chlorophenyl)-dibenzo|"b,f1[l,41thiazepine-8- carboxamide
Figure imgf000146_0002
[0465] 12.8 mg, UV/MS purity 100/100
Series I
[0466] The amidoimidoyl chlorides (Examples 262 - 271) were synthesized according to the general procedure for amide formation using 11-Chloro- dibenzo[ό,/][l,4]thiazepine-8-carbonyl chloride (300 mg, 1 mmol) and the proper amine (3 mmol) except that the reaction mixture was passed through a pad of acidic alumina oxide and eluted with a mixture of CH2Cl2 and EtOAc. The eluents were concentrated at reduced pressure and the obtained crude products were directly used in the next reactions without further purifications or characterization. Examples 262-271
Figure imgf000147_0001
Series J Examples 272-301
[0467] Examples 272-301 are prepared according to the general procedure for the synthesis of amidines starting from 15 mg of the appropriate amidoimidoyl chloride (represented by titled compounds in Examples 262 - 271) and the appropriate amine (excess), except that purification is performed by eluting (EtOAc) the products through a pad of silica. The eluents are concentrated at reduced pressure to give the crude products, which are purified by preparative HPLC/MS. Yield is determined by weighing and purity by analytical LCVMS).
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Series K Examples 302- 391
[0468] Examples 302-391 are prepared according to the general procedure for Negishi cross-coupling starting from 10-15 mg of the appropriate amidoimidoyl chloride (represented by titled compounds in Examples 262 - 271) and the proper arylzinc halide (8eq) in THF. Ammonium chloride (0.02 ml) is added to the reaction mixtures, which are then passed through a short column (Na2SO4/silica) using EtOAc as eluent. The eluents are concentrated at reduced pressure and the crude products are purified by preparative LC/MS. Yields are determined by weighing and purities by analytical LC/MS.
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000154_0002
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000157_0002
Figure imgf000158_0001
Figure imgf000158_0002
Figure imgf000158_0003
Figure imgf000159_0001
Figure imgf000159_0002
Figure imgf000160_0001
Figure imgf000160_0002
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000162_0002
Figure imgf000163_0001
Figure imgf000163_0002
Figure imgf000164_0001
Figure imgf000164_0002
Figure imgf000164_0003
Figure imgf000165_0001
Figure imgf000165_0002
Figure imgf000166_0001
Figure imgf000166_0002
Figure imgf000167_0001
Figure imgf000167_0002
Figure imgf000168_0001
Figure imgf000168_0002
Figure imgf000169_0001
Figure imgf000169_0002
Figure imgf000170_0001
Figure imgf000170_0002
Series L
[0469] The amidoimidoyl chlorides (Examples 392-403) were synthesized according to the general procedure for amide formation using 11-chloro- dibenzo[fr,/J[l,4]thiazepine-8-carbonyl chloride (300 mg, 1 mmol) and the proper amine (2.5 mmol) except that the reaction mixture was passed through a pad of silica and eluted with a mixture of THF and EtOAc. The eluents were concentrated at reduced pressure and the obtained crude products were directly used in the next reactions without further purifications or characterization.
Figure imgf000171_0001
Figure imgf000171_0002
Figure imgf000171_0003
Figure imgf000171_0004
Figure imgf000171_0005
Figure imgf000172_0001
Series M Examples 404 - 499
[0470] Examples 404-499 are prepared according to the general procedure for Negishi cross-coupling starting from 10-15 mg of the appropriate amidoimidoyl chloride (represented by title compounds in Examples 392-403) and the proper arylzinc halide (8eq) in THF.
Figure imgf000173_0001
Figure imgf000173_0003
Figure imgf000173_0002
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
[0471] The following compounds (Examples 500 - 533) were synthesised from l l-chloro-dibenzo[ό,/J[l,4] thiazepine-8-carbonyl chloride according to the general procedure for amide formation using the proper amide followed by the general procedure- for palladium catalyzed Negishi cross-coupling of amidoimidoyl chlorides and arylzinc halides or the general procedure for synthesis of ami dines. Example 500: TE)-I l-r5-chlorothiophen-2-yl)-N-propyldibenzo|"b,f][l,4]thiazepine-8- carboxamide
Figure imgf000185_0001
[0472] Amount isolated: 2.5 mg. LCMS m/z [M+H]+: 413, purity (UV/MS): 100/98, /R = 5.60 rain.
Example 501 : (Z)-11 -f4-chloro-2-fluorophenγl)-N-isobutyldibenzo['b,f|[l ,4]thiazepine-8- carboxamide
Figure imgf000185_0002
[0473] Amount isolated: 88 mg (28 %).
[0474] 1H NMR (400 MHz, CDCl3) δ 7.90 (t, IH, J= 8.4, ArH), 7.67 (t, IH, J = 0.9, ArH), 7.55 - 7.51 (m, 2H, ArH), 7.41 (dt, IH, J= 1.6, 7.6, ArH)5 7.32 - 7.24 (m, 3H3 ArH), 7.14 - 7.08 (m, 2H, ArH), 6.12 (br s, IH, NH), 3;28 (t, 2H, J= 6.8, NCH2), 1.87 (sept, IH, J = 6.8, CH1Bu), 0.97 (d, 6H, J = 6.8, 2 x CH3). LCMS m/z [M+ 1]+: 439, purity (UV/MS): 100/95, tκ = 5.63 min.
Example 502: (E)-I l-(5-chlorothiophen-2-yl)-N-isobutyldibenzo[b,f][l,41thiazepine-8- carboxamide
Figure imgf000186_0001
[0475] Amount isolated: 27 mg (15 %).
[0476] 1H NMR (400 MHz, CDCl3) δ 7.60 - 7.34 (m, 7H5 ArH)5 6.94 (d, IH, J = 4.O5 thiophenH), 6.89 (d, IH5 J = 4.0, thiopheneH), 6.15 (br m, IH5 NH), 3.26 (dd5 2H, J = 6.4, 7.2, CH2(Bu), 1-87 (m, IH, CHjBu), 0.96 (d, 6H, J = 6.8, 2 x CH3). 13C NMR (100 MHz, CDCl3) δ 166.8, 162.7, 148.5, 145.1, 140.5, 137.1, 136.2, 135.3, 133.0, 132.8, 132.1, 132.0, 131.9, 130.3, 128.4, 127.3, 124.7, 123.9, 47.6, 28.8, 20.4. LCMS m/z [M+l]+: 427, purity (UVZMS): 66/98, tR = 5.83 min.
Example 503: (EVN-butyl-1 l-(5-chlorothiophen-2-yl)dibenzo[b,f|rL4]thiazepine-8- carboxamide
Figure imgf000186_0002
[0477] Amount isolated: 1.2 mg. LCMS m/z [M+H]+: 427, purity (UV/MS): 100/86, tR = 5.96 min.
Example 504: (E)-N-O -chlorobenzy D- 11 -(4-fluoropiperidin- 1 vDdibenzo [b,fl 11 ,41thiazepine -8 -carboxamide
Figure imgf000187_0001
[0478] Amount isolated: 8 mg. LCMS m/z [M+H]+: 480, tR = 5.23 min.
Example 505: (Z)-N-(azepan-l-vO-l l-(3-chlorophenyl)dibenzorb,f][L41thiazepine-8- carboxamide
Figure imgf000187_0002
[0479] Amount isolated: 2.5 mg. LCMS m/z [M+H]+: 462, purity (UV/MS): 100/94, tR= 5.23 min.
Example 506: rZVN-rr2S,6RV2.6-dimethylpiperidin-l-ylVl l-(3- fluorophenyl)dibenzorb,firL41thiazepine-8-carboxamide
Figure imgf000187_0003
[0480] Amount isolated: 4.1 mg. LCMS m/z [M+H]+: 460, purity (UV/MS):100/61, tR = 4.89 min. Example 507: (Z)-I l-f3Λ-dichloroρhenyl)-N-(f2S,6R)-2,6-dimethylpiperidin-l- yl)dibenzo[b,f|["l,41thiazepine-8-carboxamide
Figure imgf000188_0001
[0481] Amount isolated: 0.7 mg. LCMS m/z [M+H]+: 510, purity (UV/MS): 100/100, /R = 5.68 min.
Example 508: (Ε)-N-isobutyl-l l-r3-methylthiophen-2-vπdibenzop3,f)[l,41thiazepine-8- carboxamide
Figure imgf000188_0002
[0482] Amount isolated: 9.1 mg. LCMS m/z [M+HJ+: 407, purity (UV/MS): 100/98, tR = 9.55 min.
Example 509: (Z>N-f3-chlorophenethyr)-l l-(3-chlorophenyl)dibenzofb,f|["l,41thiazepine-8- carboxamide
Figure imgf000188_0003
[0483] Amount isolated: 10 mg. LCMS m/z [M+H]+ 587, tκ = 6.28 min. Example 510; (ZVl l-(4-bromophenyl)-N-isobutyldibenzo[b.fl[1.41thiazepine-8- carboxamide
Figure imgf000189_0001
[0484] Amount isolated: 1.1 mg. LCMS m/z [M+H]+: 465, purity (UV/MS): 100/65, tR= 5.97 min.
Example 511: (Z)-N-isobuty 1- 11 -(4-methoxypheny Ddibenzo [b,f] [ 1 ,4]thiazepine- 8 - carboxamide
Figure imgf000189_0002
[0485] Amount isolated: 3.5 mg. LCMS m/z [M+H]+: 417, purity (UV/MS): 100/98, tR= 5.35 min.
Example 512: (Z)-I l-(4-fluorophenyl)-N-(piperidin-l-yl)dibenzo[b,f|JL41thiazepine-8- carboxamide
Figure imgf000189_0003
[0486] Amount isolated: 0.6 mg. LCMS m/z [M+H]+: 432, purity (UV/MS):98/93, tR = 4.41 min. Example 513: (ZVl l-(3-chlorophenyl)-N-((2S,6R)-2,6-dimethylpiperidin--l- yPdibenzo [b.flf 1 ,4]thi azepine-8 -carboxamide
Figure imgf000190_0001
[0487] Amount isolated: 10.7 mg. LCMS m/z [M+H]+: 476, purity (UV/MS): 100/54, /R = 5,24 min.
Example 514: (Z)-I l-(4-chloro-2-fluorophenvD-N-ρropyldibenzo["b,fl[l,41thiazepine-8- carboxamide
Figure imgf000190_0002
[0488] Amount isolated: 4.4 mg. LCMS m/z [M+H]+: 425, purity (UV/MS): 100/94, /R= 9.56 min.
Example 515: (EVN-butyl-1 l-(3-methylthioρhen-2-vl)dibenzorb,fiπ.41thiazeρine-8- carboxamide
Figure imgf000190_0003
[0489] Amount isolated: 6.3 mg. LCMS m/z [M+H]+: 407, purity (UV/MS): 100/100, /R = 9.63 min. Example 516: (Z)-N-(azepan-l-ylVl l-f3-fluorophenyl)dibenzorb,firL4]thiazepine-8- carboxamide
Figure imgf000191_0001
[0490] Amount isolated: 2.3 mg. LCMS m/z [M+H]+: 446, purity (UV/MS): 97/64, tκ= 4.85 min.
Example 517: (Z)-N-butyl-l l-(4-methoxyphenyl')dibenzo[b,f||'L41thiazepine-8-carboxamide
Figure imgf000191_0002
[0491] Amount isolated: 2.8 mg. LCMS m/z [M+H]+: 417, purity (UV/MS): 90/94, tκ = 5.25 min.
Example 518: (Z)-I l-(3-chlorophenylVN-(2-(pyridin-2-yl)eth.yl)dibenzoFb,firL41thiazepine- 8-carboxamide
Figure imgf000191_0003
[0492] Amount isolated: 2.6 mg. LCMS m/z [M+H]+: 470, purity (UV/MS): 100/97, tR= 4.67 min. Example 519: (E)-N-butyl- 11 -(pyridin-2-vDdibenzo Fb.fi Cl ,41thiazepine-8-carboxamide
Figure imgf000192_0001
[0493] Amount isolated: 4.1 mg. LCMS m/z [M+H]+: 388, purity (UV/MS): 100/92, tκ= 4.08 min.
Example 520: (Z)-I l-(4-methoxyphenyl)-N-propyldibenzofb,f]l"l,4"]thiazepine-8- carboxamide
Figure imgf000192_0002
[0494] Amount isolated: 0.9 mg. LCMS m/z [M+H]+: 403, purity (UV/MS): 93/100, tR= 4.95 min.
Example 521: (E)-I l-(3-methylthiophen-2-yl)-N-propyldibenzo[b,f|fl,4]thiazepine-8- carboxamide
Figure imgf000192_0003
[0495] Amount isolated: 5.7 mg. LCMS m/z [MH-H]+: 393, purity (UV/MS): 100/93, tR= 9.01 min. Example 522: (Z)-I l-(3-fluorophenyl)-N-(piperidin-l-yl)dibenzo[b,f|[l,41thiazepine-8- carboxamide
Figure imgf000193_0001
[0496] Amount isolated: 2.8 mg. LCMS m/z [M+H]+: 432, purity (UV/MS): 100/78, tR= 4.47 min.
Example 523: (Z)-N-((2S.6R)-2,6-dimethylpiperidin-l-yl)-ll-(4- fluorophenvDdibenzo fb.fi \ 1 ,41thiazepine-8-carboxamide
Figure imgf000193_0002
[0497] Amount isolated: 0.6 mg. LCMS m/z [M+H]+: 460, purity (UV/MS): 98/91, tø= 4.79 min.
Example 524: (E)-N-isopentyl-l l-(3-methylthiophen-2-yl')dibenzoP3,f|[l,41thiazepine-8- carboxamide
Figure imgf000193_0003
[0498] Amount isolated: 6.7 mg. LCMS m/z [M+H]+: 421, purity (UV/MS): 100/96, tR= 10.05 min. Example 525: (E)-I l-(5-chlorothiophen-2-ylVN-(2-methoxyethyl)dibenzorb,fiπ.41 thiazepine-8-carboxamide
Figure imgf000194_0001
[0499] Amount isolated: 5.8 mg. LCMS m/z [M+H]+: 429, purity (UVMS): 100/93, fo= 5.01 min.
Example 526: (Z)-N-isopentyl-l l-(4-methoxyphenyl)dibenzo[b,fH"l,41thiazepine-8- carboxamide
Figure imgf000194_0002
[0500] Amount isolated: 3.9 mg. LCMS m/z [M+H]+: 431, purity (UV/MS): 100/100, tR= 5.67 min.
Example 527: (E)-N-isopentyl-l l-(pγridin-2-yl)dibenzo[b,f|[l,4]thiazepine-8-carboxamide
Figure imgf000194_0003
[0501] Amount isolated: 127 mg (52 %).
[0502] 1H NMR (400 MHz, CDCl3) δ 8.69 - 8.59 (m, 2H, ArH), 8.30 - 8.25 (m, IH, ArH), 7.87 - 7.81 (m, IH, ArH), 7.71 (m, IH, ArH), 7.52 (m, 2H, ArH), 7.43 - 7.19 (m, 4H, ArH), 6.16 (br s, IH, NH), 3.48 - 3.41 (m, 2H, NCH2), 1.66 (sept, IH, J= 6.6, CH1P6n), 1.48 (q, 2H, CH2, J = 6.6), 0.93 (d, 6H, J = 6.6, 2 x CH3). LCMS m/z [M+H]+ 402, purity (UV/MS): 100/94. tκ = 4.47 min.
Example 528: (ZVl l-(4-chlorophenvπ-N-r2-(pyridin-2-vDethvπdibenzorb,firi,41thiazepine- 8-carboxamide
Figure imgf000195_0001
[0503] Amount isolated: 2.3. mg. LCMS m/z [M+H]+: 470, purity (UV/MS): 100/88, fR = 4.68 min.
Example 529: (Z)-N-fazepan-l-yD-l l-(4-fluorophenyl')dibenzo[b,f|[L4]thiazepine-8- carboxamide
Figure imgf000195_0002
[0504] Amount isolated: 0.7 mg. LCMS m/z [M+H]+: 446, purity (UV/MS): 98/92, tκ = 4.80 min.
Example 530: (E)-N-isobutyl-l l-rpyridin-2-yl)dibenzo[b,fl[L4]thiazepine-8-carboxamide
Figure imgf000195_0003
[0505] Amount isolated: 77 mg (46 %). [0506] 1H NMR (400 MHz, CDCl3) δ 8.72 - 8.70 (m, IH, ArH), 8.29 - 8.24 (m, IH, ArH), 7.87 (dt, IH5 J = 1.6, 7.6, ArH), 7.75 (m, IH, ArH), 7.57 - 7.52 (m, 3H, ArH), 7.44 - 7.38 (m, 2H, ArH)5 7.32 (dt, IH, J= 1.2, 7.6, ArH), 7.23 - 7.20 (m, IH, ArH), 6.19 (br s, IH, NH), 3.27 (t, 2H, J= 6.4, NHCH2), 1.88 (sept, IH, J= 6.4, CH/Bu), 0.97 (d, 6H, J = 6.4, 2 x CH3). LCMS m/z [M+H]+ 388, purity (UV/MS): 94/60. tκ = 4.00 min.
Example 531 : (Z)-I l-(4-bromophenylVN-propyldibenzo[b,f|fl,4]thiazepine-8-carboxamide
Figure imgf000196_0001
[0507] Amount isolated: 0.6 mg. LCMS m/z [M+H]+: 451, purity (UV/MS): 100/61, ^= 5.65 min.
Example 532: (Z)-I l-(3.4-dichlorophenylVN-(2-(pyridin-2-vnethvndibenzorb.fl[L4] thiazepine-8-carboxamide
Figure imgf000196_0002
[0508] Amount isolated: 2.1 mg. LCMS m/z [M+H]+: 504, purity (UV/MS): 100/96, tR= 5.15 min.
Example 533: (Z)-I l-(4-bromophenvl)-N-f2-methoxvethvl)dibenzorb,fiπ,41thiazepine-8- carboxamide
Figure imgf000197_0001
[0509] Amount isolated: 3.7 mg. LCMS m/z [M+H]+: 467, purity (UV/MS): 100/78, rR= 5.09 min.
Example 534: 1 l-chloro-dibenzofb^fl^lthiazepine-δ-carboxylic acid methyl ester
Figure imgf000197_0002
[0510] A mixture of the lactam (1 eq.) and PCl5 (5 eq.) in toluene was heated at 110 0C for 2 hours. The reaction mixture was then cooled to room temperature and excess of PCI5 and toluene was removed at reduced pressure using an oilpump to give crude product, which was used without further purification. The following reagents were employed: 11-oxo- 10,l l-dibenzo[b,fJ[l,4]thiazepine-8-carboxylic acid methyl ester (540 mg, 1.89 mmol), PCl5 (1.97 g, 9.47 mmol), toluene (15 mL). Purification by flash chromatography (ethyl acetate/heptane 1 :4) afforded 410 mg (71 %) of the titled compound as an yellow solid.
[0511] 1H NMR (400 MHz, CDCl3): δ 7.86 (IH5 dd, J = 2.0, 0.4Hz), 7.75 (IH, dd, J = 8.0, 1.6 Hz), 7.69-7.67 (IH, m), 7.45 (IH, dd, J = 8.4, 0.4Hz), 7.40-7.32 (3H, m), 3.82 (3H, s). 13C NMR (100 MHz, CDCl3): δ 166.2, 156.1, 146.3, 138.1, 137.9, 133.2, 133.1, 132.9, 132.4, 131.7, 130.2, 129.2, 128.1, 127.1, 52.6.
Example 535: 1 l-butyl-dibenzo|"b,f|[l,41thiazepine-8-carboxylic acid methyl ester
Figure imgf000198_0001
[0512] A flame dried 10 mL flask was charged under argon with the imidoyl chloride (1 eq.), Fe(acac)3 (5 mol%) in dry THF and cooled to - 40 "C. Functionalized arylmagnesium halide (2 eq., 1 M in THF; prepared at -40 0C) was slowly added to the solution, keeping the temperature below - 40 "C. The reaction was stirred for 5 min. at - 40 0C, then quenched with NH4CI (sat., aq.) and allowed to warm to room temperature. The resulting mixture was diluted with Et2O and the organic phase was washed with water, brine, dried (Na2SO3), filtered, and evaporated to give crude product. Purification by flash chromatography. The following reagents were employed: 11-chloro- dibenzo[b,f][l,4]thiazepine-8-carboxylic acid methyl ester (151.5 mg, 0.50 mmol), Fe(acac)3 (8.85 mg, 0.05 mmol), THF (4 mL) and N-methylpyrrolidone (0.4 mL), røButyl magnesium chloride (2 M in Et2O, 0.50 mL, 1.0 mmol). Purification by flash chromatography (ethyl acetate/heptane 1 :5) afforded 144 mg (89 %) of the titled compound as a yellow solid.
[0513] 1H NMR (400 MHz, CDCl3): δ 7.84 (IH, d, J = 1.6Hz), 7.68 (IH, dd, J = 8.0, 1.6Hz), 7.74-7.43 (2H, m), 7.40-7.31 (3H, m), 3.87 (3H, s), 3.02-2.85 (2H, m), 1.74-1.58 (2H, m), 1.55-1.41 (2H, m), 0.93 (3H, t, J = 7.2Hz). 13C NMR (100 MHz, CDCl3): δ 174.5, 166.7, 148.8, 139.7, 139.0, 134.4, 132.5, 132.3, 131.1, 130.9, 128.9, 127.9, 126.6, 126.1, 52.4, 42.2, 29.5, 22.7, 14.2.
Example 536: 1 l-butyl-dibenzo[b,f|[l,41thiazepine-8-carboxylic acid methoxy-methyl-amide
Figure imgf000198_0002
[0514] A flame dried 10 mL flask was charged under argon with the imidoyl chloride (1 eq.), Fe(acac)3 (5 mol%) in dry THF and cooled to - 40 °C. Functionalized arylmagnesium halide (2 eq., 1 M in THF; prepared at -40 "C) was slowly added to the solution, keeping the temperature below - 40 °C. The reaction was stirred for 5 min. at - 40 0C, then quenched with NH4Cl (sat., aq.) and allowed to warm to room temperature. The resulting mixture was diluted with Et2O and the organic phase was washed with water, brine, dried (Na2SO3), filtered, and evaporated to give crude product. Purification by flash chromatography. The following reagents were employed: 11-chloro- dibenzo[b,fJ[l,4]thiazepine-8-carboxylic acid methoxy-methyl-amide (61.5 mg, 0.19 mmol), Fe(acac)3 (3.53 mg, 0.001 mmol), THF (2 mL) and N-methylpyrrolidone (0.20 niL), π-Butyl magnesium chloride (2 M in Et2O, 0.11 mL, 0.23 mmol). Purification by flash chromatography (ethyl acetate/heptane 1:1) afforded 47 mg (70 %) of the titled compound as a yellow oil.
[0515] 1H NMR (400 MHz, CDCl3): δ 7.45-7.42 (3H, m), 7.39-7.29 (4H, m), 3.54 (3H,s), 3.32 (3H,s), 3.01-2.82 (2H, m), 1.69-1.59 (2H, m), 1.51-1.41 (2H, m), 0.92 (3 H, t, J = 7.2Hz). 13C NMR (100 MHz, CDCl3): δ 174.4, 169.2, 148.7, 140.0, 139.0, 135.2, 132.2, 132.1, 131.6, 130.8, 128.7, 127.9, 125.1, 124.9, 61.4, 42.2, 34.1, 29.6, 22.7, 14.1.
Example 537: (1 l-butyl-dibenzo[b,f|[l ,4]thiazepine-8-yl)-cvclohexyl-methanone
Figure imgf000199_0001
[0516] A flame dried 10 mL flask was charged under argon with 11-butyl- dibenzo[b,f][l,4]thiazepine-8-carboxylic acid methoxy-methyl-amide (29 mg, 0.08 mmol) in dry THF (2 mL) and cyclohexyl magnesium chloride (2 M in Et2O, 0.12 mL, 0.24 mmol) was then added. The resulting reaction mixture was stirred at room temperature for 1 hour and was then diluted with ether. The organic phase was washed with water, brine, dried (Na2SO3), filtered and evaporated to give crude product. Purification by prepatory TLC (ethyl acetate/heptane 1: 10) afforded 5 mg (17 %) of the titled compound as a colorless oil.
[0517] 1H NMR (400 MHz, CDCl3): δ 7.70 (IH, d, J = 2Hz), 7.60 (IH, dd, J = 8.0, 2.0Hz), 7.49-7.44 (2H, m), 7.41-7.33 (3H,m), 3.19 (IH, tt, J = 11.2, 3.2Hz), 3.04-2.97 (IH, m), 2.92-2.84 (IH, m), 1.83-1.79 (3H, m), 1.72-1.62 (3H, m), 1.51-1.21 (8H, m), 0.93 (3H, t, J = 7.6Hz). 13C NMR (100 MHz, CDCl3): δ 203.4, 174.7, 148.9, 139.8, 138.9, 137.3, 134.2, 132.8, 132.3, 130.9, 128.9, 127.9, 125.3, 124.9, 45.8, 42.3, 29.6, 29.5, 26.1, 26.0, 22.7, 14.2.
Example 538: 1-d l-chloro-dibenzorb,fiπ,41thiazepine-8-yl)-pentan-l-one
Figure imgf000200_0001
[0518] A flame dried 10 mL flask was charged under argon with 11-chloro- dibenzo[b,f][l,4]thiazepine-8-carboxylic acid methoxy-methyl-amide (34 mg, 0.10 mmol) in dry THF (2 mL) and nButyl magnesium chloride (2 M in Et2O, 0.10 mL, 0.2 mmol) was then added. The resulting reaction mixture was stirred at room temperature for 1 hour and was then diluted with ether. The organic phase was washed with water, brine, dried (Na2SO3), filtered and evaporated to give crude product. Purification by flash chromatography (ethyl acetate/heptane 1 :5) afforded 26.0 mg (81 %) of the titled compound as a yellow oil.
[0519] 1U NMR (400 MHz, CDCl3): δ 7.82 (IH, d, J = 1.6Hz), 7.77-7.74 (2H, m), 7.53 (IH, d, J = 8.4Hz), 7.47-7.39 (3H, m), 2.90 (2H, t, J = 7.2Hz), 1.68 (2H, quintet, J = 7.2Hz), 1.37 (2H, sextet, J = 7.2Hz), 0.93 (3H, t, J = 7.2Hz). 13C NMR (100 MHz, CDCl3): δ 199.5, 156.2, 146.4, 138.3, 138.1, 137.8, 133.2, 133.1(2), 132,5, 1302, 129.2, 126.6, 125.8, 38.7, 26.5, 22.6, 14.1.
Example 539: l-d l-cyclohexyl-dibenzo rbj1Fl,41 thiazepine-8-yl)-pentan-l-one
Figure imgf000200_0002
[0520] A flame dried 10 mL flask was charged under argon with the imidoyl chloride (1 eq.), Fe(acac)3 (5 mol%) in dry THF and cooled to - 40 °C. Functionalized arylmagnesium halide (2 eq., 1 M in THF; prepared at -40 °C) was slowly added to the solution, keeping the temperature below - 40 °C. The reaction was stirred for 5 min. at - 40 °C, then quenched with NH4Cl (sat., aq.) and allowed to warm to room temperature. The resulting mixture was diluted with Et2O and the organic phase was washed with water, brine, dried (Na2SO3), filtered, and evaporated to give crude product. Purification by flash chromatography. The following reagents were employed: l-(l l-chloro- dibenzo[b,fJ[l,4]thiazepine-8-yl)-pentan-l-one (26.0 mg, 0.08 mmol), Fe(acac)3 (1.41 mg, 0.004 mmol), THF (2 mL) and N-methylpyrrolidone (0.20 mL), cyclohexyl magnesium chloride (2 M in Et2O, 0.08 mL, 0.16 mmol). Purification by prep. TLC (ethyl acetate/heptane 1: 10) afforded 17.2 mg (57%) of the titled compound as an colorless oil.
[0521] 1H NMR (400 MHz, CDCl3): δ 7.71 (IH, d, J = 1.6Hz), 7.59 (IH, dd, J = 8.0, 2.0Hz), 7.48-7.43 (2H, m), 7.40-7.29 (3H, m), 2.92-2.85 (3H, m), 2.21-2.17 (IH, m), 1.98-1.93 (IH, m), 1.82-1.63 (6H, m), 1.43-1.26 (6H, m), 0.92 (3H, t, J = 7.2Hz). 13C NMR (100 MHz, CDCl3): δ 200.1, 177.8, 149.0, 140.1, 139.2, 137.9, 134.3, 132.6, 132.0, 130.6, 128.9, 127.4, 125.2, 124.3, 49.1, 38.6, 32.6, 30.2, 30.0, 26.6, 26.4, 26.1, 22.6, 14.1.
Example 540: 11 -(4-fluorophenyl>N-(thiophen-2-ylmethyl)dibenzo[b,f| [1 ,4]thiazepine-8- carboxamide
Figure imgf000201_0001
[0522] Amount isolated: 0.8 mg. LCMS m/z [M+H]+: 444, purity (UV/MS): 100/62, fR = 4.97 min.
Example 541 : 11 -(5-chlorothior)hen-2-ylVN-(thiophen-2-ylmethvndibenzo[b.fl [1.41 thiazepine-8-carboxamide
Figure imgf000201_0002
[0523] Amount isolated: 1.1 mg. LCMS m/z [M+H]+: 466, purity (UV/MS): 99/31, tR = 3.00 min. Example 542: 1 l-(3-chlorophenyl)-N-(thiophen-2-ylmethyl)dibenzo[b.f|[l,4]thiazepine-8- carboxamide
Figure imgf000202_0001
[0524] Amount isolated: 4.0 mg. LCMS m/z [M+H]+: 460, purity (UV/MS): 99/34, fo = 5.35 min.
Example 543: 1 l-(4-chlorophenyl)-N-(thiophen-2-ylmethyl)dibenzo[b,f][l,4]thiazepine-8- carboxamide
Figure imgf000202_0002
[0525] Amount isolated: 0.9 mg. LCMS m/z [M+H]+: 460, purity (UV/MS): 100/43, tR = 5.35 min.
Example 544: 1 l-(3-methylthiophen-2-yl)-N-(thiophen-2-ylmethvDdibenzo[b.firi,4] thiazepine-8-carboxamide
Figure imgf000202_0003
[0526] Amount isolated: 3.6 mg. LCMS m/z [M+H]+: 446, purity (UV/MS): 100/49, tR = 4.93 min.
Example 545 : 11 -(3 ,4-dichlorophenyl)-N-(pyridin-3-ylmethyl)dibenzo[b,f) [1 ,41thiazepine-8- carboxamide
Figure imgf000203_0001
[0527] Amount isolated: 1.5 mg. LCMS m/z [M+H]+: 489, purity (UV/MS): 96/25, fe = 4.93 min.
Example 546: 1 l-(4-chlorophenyl)-N-(furan-2-ylmethyl)dibenzo[b,f|[l,41thiazepine-8- carboxamide
Figure imgf000203_0002
[0528] Amount isolated: 4.7 mg. LCMS m/z [M+H]+: 444, purity (UV/MS): 100/58, fo = 5.19 min.
Example 547: 1 l-(4-fluorophenyl)-N-(furan-2-ylmethyl)dibenzo[b,f1[l,4]thiazepine-8- carboxamide
Figure imgf000203_0003
[0529] Amount isolated: 1.7 mg. LCMS m/z [M+H]+: 428, purity (UV/MS): 99/48, tR = 4.73 min.
Example 548: 1 l-(5-chlorothiophen-2-yl)-N-(furan-2-ylmethyl)dibenzorb,firL41thiazepine- 8-carboxamide
Figure imgf000204_0001
[0530] Amount isolated: 6.3 mg. LCMS m/z [M+H]+: 450, purity (UV/MS): 100/37, tκ = 5.21 min.
Example 549: 1 l-(3-fluorophenyl)-N-rthiophen-2-ylmethvDdibenzofb,f][l,41thiazepine-8- carboxamide
Figure imgf000204_0002
[0531] Amount isolated: 3.7 mg. LCMS m/z [M+H]+: 444, purity (UV/MS): 100/47, /R = 5.07 min.
Example 550: 1 l-f3,4-dichlorophenylVN-(furan-2-ylmethyl)dibenzo['b,f|[L4'|thiazepine-8- carboxamide
Figure imgf000204_0003
[0532] Amount isolated: 9.4 mg. LCMS m/z [M+H]+: 478, purity (UV/MS): 100/62, tκ = 5.55 min.
Example 551 : 1 l-r3.4-dichlorophenyl)-N-(2-(pyridin-3-yl)ethyl)dibenzorb,firi,41thiazepine- 8-carboxami
Figure imgf000205_0001
[0533] Amount isolated: 4.8 mg. LCMS m/z [M+H]+: 503, purity (UVMS): 100/34, tR = 4.93 min.
Example 552: 1 l-(3-chlorophenyl)-N-(furan-2-ylmethyl)dibenzo[b,f|[l,4]thiazepine-8- carboxamid
Figure imgf000205_0002
[0534] Amount isolated: 7.2 mg. LCMS m/z [M+H]+: 444, purity (UV/MS): 100/70, tR = 5.13 min.
Example 553: 1 l-(5-chlorothiophen-2-ylVN-(2-(pyridin-3-vDethvndibenzorb,f|[l,4] thiazepine-8-carboxamide
Figure imgf000205_0003
[0535] Amount isolated: 5.9 mg. LCMS m/z [M+H]+: 475, purity (UV/MS): 97/17, rR = 4.52 min.
Example 554: 1 l-(3,4-dichlorophenyl)-N-(2-(pyridin-4-vDethvDdibenzo[b,f|[l,41thiazepine- 8-carboxamide
[0536] Amount isolated: 0.8 mg. LCMS m/z [M+H]+: 504, purity (UV/MS): 97/44, tR = 4.90 min.
Example 555: 1 l-(5-chlorotMophen-2-ylVN-(2-fρyridin-4-yl)ethvndibeDzorb.firi,41 thiazepine-8-carboxamide
Figure imgf000206_0002
[0537] Amount isolated: 1.8 mg. LCMS m/z [M+H]+: 475, purity (UV/MS): 100/70, tR = 4.52 min.
Example 556: 1 l-(4-fluorophenylVN-(pyridin-3-ylmethyl)dibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000206_0003
[0538] Amount isolated: 1.3 mg. LCMS m/z [M+H]+: 439, purity (UV/MS): 99/47, tR = 4.05 min.
Example 557: 1 l-(4-fluorophenylVN-(2-(pyridin-3 -vDethvndibenzorb.fi Fl.41thiazepine-8- carboxamide
Figure imgf000207_0001
[0539] Amount isolated: 1.3 mg. LCMS m/z [M+H]+: 453, purity (UV/MS): 98/38, tR = 4.07 min.
Example 558: 11 -(3 -fluorophenyl)-N-(pyridin-3 -ylmethyPdibenzo [b.f| [ 1 ,4]thiazepine-8- carboxamide
Figure imgf000207_0002
[0540] Amount isolated: 3.0 mg. LCMS m/z [M+H]+: 439, purity (UV/MS): 99/40, /R = 4.10 min.
Example 559: 1 l-r4-fluorophenylVN-r2-('pyridin-4-yl')ethyl)dibenzorb,f]ri.41thiazepine-8- carboxamide
Figure imgf000207_0003
[0541] Amount isolated: 0.9 mg. LCMS m/z [M+H]+: 453, purity (UV/MS): 89/34, tR = 4.07 min.
Example 560: 1 l-(2-fluorophenyl)-N-('thiophen-2-ylmethyl)dibenzo|'b,firi,41thiazepine-8- carboxamide
Figure imgf000208_0001
[0542] Amount isolated: 3.9 mg. LCMS m/z [M+H]+: 444, purity (UV/MS): 100/41, fe = 4.67 min.
Example 561 : 1 l-(3-fluorophenyl)-N-(furan-2-ylmethyl)dibenzo|"b,fl[l,41thiazepine-8- carboxamide
Figure imgf000208_0002
[0543] Amount isolated: 5.5 mg. LCMS m/z [M+H]+: 428, purity (UV/MS): 100/47, fo = 4.75 min.
Example 562: N-ffuran-2-ylmethyl)-l l-(3-methylthiophen-2-yl)dibenzo[b,f|[L41thiazepine- 8-carboxamide
Figure imgf000208_0003
[0544] Amount isolated: 7.9 mg. LCMS m/z [M+H]+: 430, purity (UV/MS): 95/55, tR = 4.70 min.
Example 563: 1 l-(3-chloroρhenyl)-N-(2-rpyridin-3-yl)ethyl)dibenzorb,firi,41thiazepine-8- carboxamide
Figure imgf000209_0001
[0545] Amount isolated: 6.2 mg. LCMS m/z [M+H]+: 469, purity (UVMS): 100/64, tR = 4.50 min.
Example 564: 1 l-(2-fluorophenyl)-N-(pyridin-3-ylmethyl)dibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000209_0002
[0546] Amount isolated: 4.0 mg. LCMS m/z [M+H]+: 439, purity (UV/MS): 99/39, rR = 3.75 min.
Example 565: 1 l-^-chlorophenylVN-fΣ-Cpyridin-S-vDethvDdibenzorb^fifl^lthiazepine-δ- carboxamide
Figure imgf000209_0003
[0547] Amount isolated: 5.6 mg. LCMS m/z [M+H]+: 469, purity (UV/MS): 88/19, rR = 4.50 min.
Example 566: 1 l-(3-fluorophenyl')-N-(2-fpyridin-3-yl>>ethvπdibenzorb.firi.41thiazepine-8- carboxamide
Figure imgf000210_0001
[0548] Amount isolated: 4.4 mg. LCMS m/z [M+H]+: 453, purity (UV/MS): 100/39, tκ = 4.12 min.
Example 567: 11 -f4-ChlorophenviyN-f2-fpyridine-4-vDethyl)dibenzo[b,fl rh41thiazepine-8- carboxamide
Figure imgf000210_0002
[0549] Amount isolated: 2.5 mg. LCMS m/z [M+H]+: 469, purity (UV/MS): 97/37, tR = 4.47 min.
Example 568: 1 l-(pyridin-2-yl)-N-(thiophen-2-ylmethyl)dibenzorb,f1[l,41thiazepine-8- carboxamide
Figure imgf000210_0003
[0550] Amount isolated: 2.1 mg. LCMS m/z [M+H]+: 427, purity (UV/MS): 100/68, rR = 3.88 min. Example 569: 1 l-(3-FluorophenylVN-f2-(pyridin-4-yl)ethyl)dibenzo[b,f|[l,41thiazepine-8- carboxamide
Figure imgf000211_0001
[0551] Amount isolated: 3.7 mg. LCMS m/z [M+H]+: 453, purity (UV/MS): 94/35, fa = 4,08 min.
Example 570: 1 l-(2-FluorophenylVN-('furan-2-ylmethyl)dibenzorb,firi,41thiazepine-8- carboxamide
Figure imgf000211_0002
[0552] Amount isolated: 5.7 mg. LCMS m/z [M+H]+: 428, purity (UV/MS): 94/34, tκ = 4.45 min.
Example 571 : 1 l-r3-methylthiophen-2-vn-N-r2-(pyridin-3-vDethvndibenzorb.firi.41 thiazepine-8-carboxamide
Figure imgf000211_0003
[0553] Amount isolated: 3.3 mg. LCMS m/z [M+H]+: 455, purity (UV/MS): 100/42, tR = 4.02 min.
Example 572: 1 l-(2-fluorophenylVN-(2-(pyridin-3-vDethvπdibenzorb,firi.41thiazepine-8- carboxamide
Figure imgf000212_0001
[0554] Amount isolated: 6.3 mg. LCMS m/z [MH-H]+: 453, purity (UV/MS): 95/34, ^R = 3.78 min.
Example 573: N-C furan-2-y lmethylV 11 -f pyridin-2-vDdibenzo Fb.fl [ 1 ,41thiazepine-8- carboxamide
Figure imgf000212_0002
[0555] Amount isolated: 3.1 mg. LCMS m/z [M+H]+: 411, purity (UV/MS): 100/58, tR = 3.62 min.
Example 574: 11 -O-metfayl1faiophen-2-ylVN-fpyridin-3-ylmethvndiber-zorb.fi f 1,41 thiazepine-8-carboxamide
Figure imgf000212_0003
[0556] Amount isolated: 2.2 mg. LCMS m/z [M+H]+: 441, purity (UV/MS):100/35, tR = 3.98 min.
Example 575: 1 l-f3-methylpyridin-2-yl)-N-fthiophen-2-ylmethyl)dibenzorb,fl|"l,41 thiazepine-8-carboxamide
Figure imgf000212_0004
[0557] Amount isolated: 1.6 mg, LCMS m/z [M+H]+: 441, purity (UV/MS): 100/54, tκ = 3.85 min.
Example 576: 11 -(3-methylthiophen-2-ylVN-(2-(pyridin-4-yl)ethyl)dibenzorb.f1 Tl .41 thiazepine-8-carboxamide
Figure imgf000213_0001
[0558] Amount isolated: 1.8 mg. LCMS m/z [M+H]+: 455, purity (UV/MS): 98/22, tR = 4.00 min.
Example 577: 1 l-(3-fluorophenyl)-N-(pyridin-4-ylmethyl)dibenzo[b,f)[l,41thiazepine-8- carboxamide
Figure imgf000213_0002
[0559] Amount isolated: 2.4 mg. LCMS m/z [M+H]+: 439, purity (UV/MS): 98/35, tR = 4.03 min.
Example 578: 1 l-ΩΛ-dichlorophenylVN-fΣ-Cpyridin-S-vDethvDdibenzofb.firiΛlthiazepine- 8-carboxamide
Figure imgf000213_0003
[0560] Amount isolated: 3.9 mg. LCMS m/z [M+H]+: 503, purity (UV/MS): 96/24, tR = 4.58 min. Example 579: 1 l-(2-fluorophenyl)-N-(pyridin-4-ylmethvπdiben2θ[b,fH"l,41thiazepine-8- carboxamide
Figure imgf000214_0001
[0561] Amount isolated: 2.5 mg. LCMS m/z [M+H]+: 439, purity (UV/MS): 100/50, tR = 3.72 min.
Example 580: 1 l-(2-chlorophenyl)-N-(thiophen-2-ylmethyl)dibenzo[b,f]ri,41thiazepine-8- carboxamide
Figure imgf000214_0002
[0562] Amount isolated: 1.4 mg. LCMS m/z [M+H]+: 460, purity (UV/MS): 99/51, tR = 4.88 min.
Example 581: 1 l-(2-chlorophenyl)-N-(furan-2-ylmethyl)dibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000214_0003
[0563] Amount isolated: 6.2 mg. LCMS m/z [M+H]+: 444, purity (UV/MS): 100/34, tR = 4.65 min.
Example 582: 1 l-r3-methylthiophen-2-vn-N-(pyridin-4-ylmethvndibenzorb,f|[1.41 thiazepine-8-carboxamide
Figure imgf000215_0001
[0564] Amount isolated: 1.6 mg. LCMS m/z [M+H]+: 441, purity (UV/MS): 99/41, fo = 3.97 min.
Example 583: 1 l-(2-chlorophenyl)-N-(2-rpyridin-3-vnethyl)dibenzorb,fiπ,41thiazepine-8- carboxamide
Figure imgf000215_0002
[0565] Amount isolated: 4.5 mg. LCMS m/z [M+H]+: 470, purity (UV/MS): 100/40, tR = 3.97 min.
Example 584: 1 l-(ρyridin-2-ylVN-('2-(pyridin-4-vDethylMibenzorb.firi,41thiazepine-8- carboxamide
Figure imgf000215_0003
[0566] Amount isolated: 3.0 mg. LCMS m/z [M+H]+: 436, purity (UV/MS): 98/60, fa = 2.90 min.
Example 585: 1 l-fΣ^-dichlorophenylVN-fΣ-rpyridin^-vnethvndibenzoCb.firiΛithiazepine- 8-carboxamide
Figure imgf000216_0001
[0567] Amount isolated: 1.2 mg. LCMS m/z [M+H]+: 503, purity (UV/MS): 96/30, tκ = 4.58 min.
Example 586: 1 l-f2-chlorophenylVN-('2-fpyridin-4-vnethyl)dibenzorb,firL41thiazeρine-8- carboxamjde
Figure imgf000216_0002
[0568] Amount isolated: 2.4 mg. LCMS m/z [M+H]+: 469, purity (UV/MS): 93/36, tR = 3.95 min.
Example 587: 1 l-(3-methylpyridin-2-yl)-N-(pyridin-3-ylmethyl)dibenzo[b,f|[l,4]thiazepine- 8-carboxamide
Figure imgf000216_0003
[0569] Amount isolated: 3.5 mg. LCMS m/z [M+H]+: 436, purity (UV/MS): 92/7 MR = .93 min.
Example 588: 1 l-O-fluorophenylVN-CS-methylisoxazol-S-vDdibenzoCb^fin^ithiazepine-S- carboxamide
Figure imgf000217_0001
[0570] Amount isolated: 0.6 mg. LCMS m/z [M+H]+: 430, purity (UV/MS): 100/36, tR = 4.92 min.
Example 589: 1 l-(3-methylpyridin-2-ylVN-(2-(pyridin-3-yl)ethyl')dibenzorb.f|[1.41 thiazepine-8-carboxamide
Figure imgf000217_0002
[0571] Amount isolated: 2.2 mg. LCMS m/z [M+H]+: 450, purity (UV/MS): 93/80, tR = 2.93 min.
Example 590: 1 l-(4-chlorobenzylamino)-N'-r2-phenylacetyl)dibenzo[b,f]ri,41thiazepine-8- carbohydrazide
Figure imgf000217_0003
[0572] Amount isolated: 5.1 mg. LCMS m/z [M]: 527, purity (UV/MS): 97/67, tR = 11.92 min.
Example 591 : N-(5-methylisoxazol-3-yD-l l-(pyridin-2-yl)dibenzorb,fl[1.4]thiazepine-8- carboxamide
Figure imgf000218_0001
[0573] Amount isolated: 0.6 mg. LCMS m/z [M+H]+: 412, purity (UV/MS): 97/72, fo = 3.68 min.
Example 592: 1 l-(2-fluorophenyl)-N-f5-methylisoxazol-3-yl)dibenzoP3,f|[l,41thiazepine-8- carboxamide
Figure imgf000218_0002
[0574] Amount isolated: 0.7 mg. LCMS m/z [M+H]+: 429, purity (UV/MS): 100/54, tκ = 4.59 min.
Example 593: 1 l-(4-chlorobenzylamino)-N-(pyridin-2-ylmetliyl)dibenzo[b,fl|'l,4]thiazepine- 8-carboxamide
Figure imgf000218_0003
[0575] Amount isolated: 8.6 mg. LCMS m/z [M+H]+: 485, purity (UV/MS): 95/77, ^ = 10.02 min.
Example 594: N-(5-methylisoxazol-3-yD-l l-C3-methylpyridin^2-yl)dibenzorb,fl["L41 thiazepine-8-carboxamide
Figure imgf000219_0001
[0576] Amount isolated: 0.8 mg. LCMS m/z [M+H]+: 426, purity (UV/MS): 99/60, tR = 3.72 min.
Example 595 : 11 -(4-chlorobenzylamino)-N-(2-oxoazepan-3-yl)dibenzo[b,f] [1 ,4]thiazepine- 8-carboxamide
Figure imgf000219_0002
[0577] Amount isolated: 4.7 mg. LCMS m/z [M+H]+: 505, purity (UV/MS): 88/40, fe = 11.36 min.
Example 596: N'-benzoyl-l l-(4-chlorobenzylamino)dibenzo[b,fl|"l,4]thiazepine-8- carbohydrazide
Figure imgf000219_0003
[0578] Amount isolated: 7.1 mg. LCMS m/z [M+H]+: 513, purity (UV/MS): 97/73, tR = 11.66 min.
Example 597: 1 l-r3-methylpyridin-2-yl)-N-('2-(pyridin-4-vnethyl)dibenzorb.firi.41 thiazepine-8 -carboxamide
Figure imgf000220_0001
[0579] Amount isolated: 1.0 mg. LCMS m/z [M+H]+: 450, purity (UV/MS): 88/68, te = 2.92 min.
Example 598: 1 l-('4-chlorobenzylaminoVN-methoxydibenzo[b,f][l,4]thiazepine-8- carboxamide
Figure imgf000220_0002
[0580] Amount isolated: 5.0 mg. LCMS m/z [M+H]+: 424, purity (UV/MS): 92/57, tκ = 10.82 min.
Example 599: 1 l-("2-chlorophenyl)-N-(pyridin-3-ylmethyl)dibenzo[b,f|[l,4]thiazepine-8- carboxamide
Figure imgf000220_0003
[0581] Amount isolated: 1.7 mg. LCMS m/z [M+H]+: 455, purity (UV/MS): 99/35, fo = 3.95 min.
Example 600: N-ffuran-2-ylmethylVl l-(3-methylρyridin-2-yl)dibenzorb.fiπ.41thiazepine-8- carboxamide
Figure imgf000221_0001
[0582] Amount isolated: 2.1 mg. LCMS m/z [M+H]+: 425, purity (UV/MS): 86/40, tκ = 3.65 min.
Example 601 : 1 l-rpyridin-2-ylVN-(2-rpyridin-3-yl)ethvndibenzorb,fH"1.41thiazeρine-8- carboxamide
Figure imgf000221_0002
[0583] Amount isolated: 3.6 mg. LCMS m/z [M+H]+: 436, purity (UV/MS): 96/61, tκ = 2.90 min.
Example 602: 1 l-(4-chlorobenzylamino)-N-(pyridin-3-ylmethyl)dibenzo[b,f1[l ,4]thiazepine- 8-carboxamide
Figure imgf000221_0003
[0584] Amount isolated: 8.9 mg. LCMS m/z [M+H]+: 485, purity (UV/MS): 99/100, tR = 9.87 min.
Example 603: 1 l-(2-chlorophenyl)-N-(pyridin-4-ylmethyl)dibenzorb,f)[l,4]thiazepine-8- carboxamide
Figure imgf000222_0001
[0585] Amount isolated: 1.8 mg. LCMS m/z [M+H]+: 455, purity (UV/MS): 97/39, tR = 3.92 min.
Example 604; 1 l-r4-chlorobenzylamino)-N-(pyridin-4-ylniethyl)dibenzo|"b,f1[L4]thiazepine- 8-carboxamide
Figure imgf000222_0002
[0586] Amount isolated: 4.7 mg. LCMS m/z [M+H]+: 485, purity (UV/MS): 93/95, J1 R = 9.88 min.
Example 605: 1 l-(4-chlorobenzylamino)-N-(4-sulfamoylbenzyl)dibenzorb,f|[L41thiazepine- 8-carboxamide
Figure imgf000222_0003
[0587] Amount isolated: 6.1 mg. LCMS m/z [M+H]+: 563, purity (UV/MS): 77/43, fo = 11.56 min.
Example 606: 11 -(5-Bromopyridin-2-yl)-dibenzo|"b,f] [1.4]thiazepine-8-carboxylic acid butylamide.
Figure imgf000223_0001
Preparation of the zinc reagent:
[0588] A dry flask equipped with a magnetic bar was charged with zinc dust. The reaction flask was flushed with argon and a solution of 1 ,2-dibromoethane (100 mg, 0.53 mmol) in iV,N-dimethylacetamide (1.5 mL) was added. The zinc suspension was shortly heated with a heat gun until evolution of ethylene occurred (repeated twice).
[0589] The reaction mixture was allowed to cool to room temperature. Trimethylsilyl chloride (0.30 mL, 2.3 mmol) was added in two portions. After 15 minutes stirring at room temperature a solution of 5-bromo-2-iodopyridine (1.42 g, 5.0 mmol) in JV5JV- dimethylacetamide (3.0 mL) was added to the zinc suspension at 5O0C. The reaction mixture was stirred at 700C for 3 hours. Conversion of the starting material was followed by GC using decane as the internal standard. After 3 hours at 7O0C, 60% of the starting material was converted to the desired zinc reagent. Stirring was continued overnight at 7O0C, which gave full conversion. The reaction mixture was allowed to cool to room temperature and diluted with dry THF (3.0 mL). The remaining zinc was allowed to settle. The obtained solution of 5- bromo-2-pyridylzinc iodide was used immediately in the next step.
[0590] Bis(dibenzylideneacetone)palladium (18 mg, 0.031 mmol) and tri-2- furylphosphine (15 mg, 0.065 mmol) were dissolved in dry THF (1.0 mL) in a dry flask, under argon atmosphere. A solution of 1 l-chloro-dibenzofέ./Jfl^Jthiazepine-carboxylic acid butylamide (prepared as previously described, 200 mg, 0.63 mmol) in dry THF (2.0 mL) was added to the flask. A solution of the freshly prepared 5-bromo-2-pyridylzinc iodide (3 mL, 2.0 mmol) was added dropwise to the reaction mixture at room temperature. After 20 hours stirring at room temperature the reaction mixture was partitioned between aqueous NH4Cl (sat) and EtOAc. The organic layer was dried over Na2SO4, filtered end evaporated to dryness. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 15-30% EtOAc in toluene. The isolated product was repurified using an acidic exchange cartridge eluting with NH3 in MeOH. Yield: 5.8 mg, 2%. [0591] LCMS m/z 467 [M+H]+ HPLC tR = 4.3 min. 1H NMR (CDCl3, 400 MHz) δ 8.72 (m, IH, Ar-H), 8.24 (m, IH, Ar-H), 8.01-7.98 (m, IH, Ar-H), 7.71 (m, IH, Ar-H), 7.66-7.52 (m, 3H, Ar-H), 7.43 (m, IH, Ar-H), 7.32 (m, IH, Ar-H), 7.21 (m, IH, Ar-H), 6.12- 6.03 (broad s, IH, NH), 3.45 (q, 2H, J = 7.2 Hz, CH2Bu), 1.58 (pentet, 2H, J = 7.2 Hz, CH2Bu)5 2.80 (m, 2H5 J= 7.2 Hz, CH2Bu), 0.95 (t, 3H, J= 7.2 Hz, CH3Bu).
Example 607: General procedure for the synthesis of the zinc reagents from bromopyri dines:
Figure imgf000224_0001
[0592] 2-Bromo-5-halopyridine (3 mmol) was dissolved in THF (5.5 mL) and isopropylmagnesium chloride (2 M in THF; 1.5 mL; 3.0 mmol) was added at room temperature. After 2hours, zinc bromide (1 M in THF; 3.0 mL; 3.0 mmol) was added and the mixture was stirred at room temperature under argon over night. The crude mixture was used immediately in the next step.
Example 608: 1 l-(5-Fluoropyridin-2-yl)-dibenzo[6,/|["l,41thiazepine-carboxylic acid butylamide
Figure imgf000224_0002
[0593] A reaction flask was charged with l l-chloro-dibenzo[δj][l,4]thiazepine- carboxylic acid butylamide (0.17 g; 0.50 mmol) and bis(triphenylphosphine)palladium(II) chloride (36.0 mg; 0.050 mmol) under argon. THF (5 mL) was added followed by the addition of 5-fluoro-2-pyridylzinc bromide (0.15 M in THF; 12.5 mL; 1.8 mmol) at room temperature. After 5 hours, aqueous NH4Cl (sat) was added to the mixture and extracted with EtOAc. The combined organic layers were washed with brine, dried (Na2SO,)), filtered and concentrated in vacuo. The crude mixture was purified by silica gel column chromatography (0-20% EtOAc in toluene) followed by ion exchange column chromatography (eluting with 2% NH3 in MeOH) and recrystallization from MeOH to yield the title compound as a yellow solid (54.4 mg; 27%).
[0594] LCMS m/z 406 [M+H]+, purity (UV/MS) 99/95, tκ = 8.38 min. 1H NMR (CDCl3, 400 MHz) δ 8.50 (d, IH, J= 2.8 Hz, ArH), 8.38 - 8.42 (m, IH, ArH), 7.68 (d, IH, J = 0.4 Hz, ArH), 7.50 - 7.58 (m, 4H, ArH), 7.40 - 7.44 (m, IH, ArH), 7.30 - 7.34 (m, IH, ArH), 7.20 - 7.25 (m, IH, ArH), 6.03 (br m, IH, NH), 3.44 (q, 2H, J= 6.8 Hz, CH2), 1.54 - 1.62 (m, 2H, CH2), 1.36 - 1.45 (m, 2H, CH2), 0.95 (t, 3H, J= 7.2 Hz, CH3).
Example 609: 1 l-(5-Chloropyridin-2-yl)-dibenzo[6,/][l,4]thiazepine-carboxylic acid butylamide
Figure imgf000225_0001
[0595] A reaction flask was charged with 11-chloro-dibenzo [&,/][ l,4]thiazepine- carboxylic acid butylamide (0.17 g; 0.50 mmol) and bis(triphenylphosphine)palladium(II) chloride (36.0 mg; 0.050 mmol) under argon. THF (5 mL) was added followed by the addition of 5-chloro-2-pyridylzinc bromide (0.15 M in THF; 12.5 mL; 1.8 mmol) at room temperature. After 5 hours, aqueous NH4Cl (sat) was added to the mixture and extracted with EtOAc. The combined organic layers were washed with brine, dried (Na2SO4), filtered and concentrated in vacuo. The crude mixture was purified by silica gel column chromatography (0-20% EtOAc in toluene) followed by ion exchange column chromatography (eluting with 2% NH3 in MeOH) and recrystallization from MeOH to yield the title compound as a yellow solid (69.5 mg; 33%).
[0596] LCMS m/z Ml [M+H]+, purity (UV/MS) 98/88, tR = 6.43 min. 1H NMR (CDCl3, 400 MHz) δ 8.60 (d, IH, J = 1.6 Hz, ArH), 8.31 (d, IH, J = 8.8 Hz, ArH), 7.82 - 7.85 (m, IH, ArH), 7.69 (d, IH, J= 0.4 Hz, ArH), 7.51 - 7.55 (m, 3H, ArH), 7.40 - 7.44 (m, IH, ArH), 7.32 - 7.34 (m, IH, ArH), 7.20 - 7.25 (m, IH, ArH), 6.03 (br m, IH, NH), 3.44 (q, 2H, J= 7.2 Hz, CH2), 1.54 - 1.62 (m, 2H, CH2), 1.37 - 1.47 (m, 2H, CH2), 0.95 (t, 3H, J = 7.6 Hz, CH3). Example 610: 1 l-r5-Fluoropyridin-2-ylVdibenzorά,/1[l,4]thiazepine-carboxylic acid piperidin- 1 -ylamide
Figure imgf000226_0001
[0597] A reaction flask was charged with 11-chloro-dibenzo [b,f][ 1 ,4]thiazepine- carboxylic acid piperidin- 1 -ylamide (80.0 mg; 0.22 mmol) and bis(triphenylphosphine) palladium(II)chloride (15.1 mg; 0.022 mmol) under argon. THF (3 mL) was added followed by the addition of 5-fluoro-2-pyridylzinc bromide (0.15 M in THF; 5.0 mL; 0.75 mmol) at room temperature. After 3 hours, aqueous NH4Cl (sat) was added to the mixture and extracted with EtOAc. The combined organic layers were washed with brine, dried (Na2SC^), filtered and concentrated in vacuo. The crude mixture was purified by silica gel column chromatography (0-30% EtOAc in toluene), ion exchange column chromatography (eluting with 2% NH3 in MeOH) and recrystallization from EtOAc to yield the title compound as a yellow solid (9.8 mg; 10%).
[0598] LCMS m/z 433 [M+H]+, purity (UVMS) 97/92, tκ = 3.80 min. 1H NMR (CDCl3, 400 MHz) δ 8.50 (d, IH, J= 2.8 Hz, ArH), 8.38 - 8.40 (m, IH, ArH), 7.67 (d, IH, J = 0.4 Hz, ArH), 7.51 - 7.56 (m, 4H, ArH), 7.40 - 7.44 (m, IH, ArH), 7.30 - 7.34 (m, IH, ArH), 7.20 - 7.24 (m, IH, ArH), 6.69 (br m, IH, NH), 2.82 - 2.86 (m, 4H, CH2), 1.73 - 1.79 (m, 4H, CH2), 1.44 - 1.48 (m, 2H, CH2).
Example 611: 11 -(5 -Chloropyridin-2- y IV dibenzo [b,f]\l ,4~|thiazepine-carboxy lie acid piperidin- 1 -ylamide
Figure imgf000226_0002
[0599] A reaction flask was charged with l l-chloro-dibenzo[6,/j[l,4]thiazepine- carboxylic acid piperidin- 1 -ylamide (80.0 mg; 0.22 mmol) and bis(triphenylphosphine) palladium(II)chloride (15.1 mg; 0.022 mmol) under argon. THF (3 mL) was added followed by the addition of 5-chloro-2-pyridylzinc bromide (0.15 M in THF; 5.0 mL; 0.75 mmol) at room temperature. After 3 hours, aqueous NH4Cl (sat) was added to the mixture and extracted with EtOAc. The combined organic layers were washed with brine, dried (Na2SO4), filtered and concentrated in vacuo. The crude mixture was purified by silica gel column chromatography (0-30% EtOAc in toluene) and recrystallization from EtOAc to yield the title compound as a yellow solid (13.8 mg; 14%).
[0600] LCMS m/z 449 [M+H]+, purity (UV/MS) 99/87, tR = ISA min. 1H NMR (CDCl3, 400 MHz) δ 8.60 (d, IH5 J= 1.6 Hz3 ArH), 8.301 (d, IH, J= 8.0 Hz, ArH), 7.82 - 7.84 (m, IH, ArH), 7.67 (d, IH, J= 0.4 Hz, ArH), 7.51 - 7.55 (m, 3H, ArH), 7.40 - 7.44 (m, IH, ArH), 7.30 - 7.34 (m, IH, ArH), 7.20 - 7.22 (m, IH, ArH), 6.68 (br m, IH, NH), 2.81 - 2.83 (m, 4H, CH2), 1.74 - 1.78 (m, 4H, CH2), 1.42 - 1.48 (m, 2H, CH2).
Example 612: Receptor Selection and Amplification Technology Assay
[0601] The functional receptor assay, Receptor Selection and Amplification Technology (R-SAT®), was used to investigate the pharmacological properties of known and novel CBl compounds. 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.
[0602] Briefly, NIH3T3 cells were grown in 96 well tissue culture plates to 70- 80% confluence. Cells were transfected for 16-20 h with plasmid DNAs using Polyfect (Qiagen Inc.) using the manufacturer's protocols. R-SATs were 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. AU receptor constructs used were in the pSI- derived mammalian expression vector (Promega Inc). The CBl receptor gene was amplified by PCR from genomic DNA using oligodeoxynucleotide primers based on the published sequence (GenBank Accession # X54937) SEQ ID NO: 1 encodes a CBl receptor truncated after amino acid 417 (SEQ ID NO: 2). The CB2 gene was cloned by performing a PCR reaction on mRNA from spleen. The PCR product containing the entire coding sequence of the CB2 gene was cloned into an expression vector such that the CB2 gene was 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 were transfected for 16-20 h, then trypsinized and frozen in DMSO. Frozen cells were later thawed, plated at -10,000 cells per well of a 96 half-area well plate that contained drug. With both methods, cells were then grown in a humidified atmosphere with 5% ambient CO2 for five days. Media was then removed from the plates and marker gene activity was measured by the addition of the β-galactosidase substrate o-nitrophenyl β-D-galactopyranoside (ONPG) in PBS with 0.5% NP-40. The resulting colorimetric reaction was measured using a spectrophotometric plate reader (Titertek Inc.) at 420 nm. All data was analyzed using the XLFit (IDBSm) computer program. pIC5o represents the negative logarithm of the concentration of ligand that caused 50% inhibition of the constitutive receptor response. Percent inhibition was 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%.
[0603] These experiments provide a molecular profile, or fingerprint, for each of these agents at the human CBl receptor. As can be seen in Table 1, the compounds are inverse agonists at the CB 1 receptor. Additional pICso data shown in Appendix A.
TABLE 1
Figure imgf000228_0001
[0604] It will be appreciated that the foregoing assay may be used to identify compounds which are agonists, inverse agonists or antogonists 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 CB 1 receptor or a truncated CB 1 receptor of SEQ ID NO: 2.
[0605] Using the following methods, the compounds disclosed herein were evaluated for their ability to bind to a CBl receptor. The compounds were tested using a receptor binding assay and then determining of any change in GTPgamma S binding of transfected cells.
Example 613: CBl Receptor Binding Assays
[0606] 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 was examined in rat brain membrane preparations as follows.
[0607] Membrane preparations - Whole brains were harvested from Harlan Sprague Dawley rats and placed in 50 ml Falcon Tubes on ice. The volume was made up to 30 ml with ice-cold membrane buffer (20 mM HEPES, 6 mM MgCl2, 1 mM EDTA, pH 7.2). The Brains were homogenized with a Brinkmann Polytron PT3000 at 20,000 rpm for 40 s. The homogenate was spun at 1,000 x g for 10 min at 4°C to remove nuclei and cellular debris. The supernatant was collected and re-centrifuged as previously before membranes were 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 -8O0C.
[0608] Membrane Binding - 10 μg of membranes were 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 were 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 were dried under a heat lamp before addition of 50 μl of scintillation fluid to each well (Microscint 20, Packard, Shelton, CT). Plates were counted on a Topcount NXT (Packard, Shelton, CT).
[0609] Data Analysis - Graphs were plotted and KD values were determined by nonlinear regression analysis using Prism software (GraphPad version 4.0, San Diego, CA, USA).
Table 2. Binding of CBl antagonists to native CBl receptors
[0610] These results demonstrate that the compounds described herein bind with high affinity to native CBl receptors.
Figure imgf000230_0001
[0611] It will be appreciated that the CBl receptor binding assay of the foregoing example 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.
Example 614: Sequences for truncated CBl receptors
[0612] Below are sequences encoding a truncated CB 1 receptor.
SEQ ID NO:! :
ATGAAGTCGATCCTAGATGGCCTTGCAGATACCACCTTCCGCACCATCACCACTG
ACCTCCTGTACGTGGGCTCAAATGACATTCAGTACGAAGACATCAAAGGTGACA
TGGCATCCAAATTAGGGTACTTCCCACAGAAATTCCCTTTAACTTCCTTTAGGGG
AAGTCCCTTCCAAGAGAAGATGACTGCGGGAGACAACCCCCAGCTAGTCCCAGC
AGACCAGGTGAACATTACAGAATTTTACAACAAGTCTCTCTCGTCCTTCAAGGAG
AATGAGGAGAACATCCAGTGTGGGGAGAACTTCATGGACATAGAGTGTTTCATG
GTCCTGAACCCCAGCCAGCAGCTGGCCATTGCAGTCCTGTCCCTCACGCTGGGCA
CCTTCACGGTCCTGGAGAACCTCCTGGTGCTGTGCGTCATCCTCCACTCCCGCAG
CCTCCGCTGCAGGCCTTCCTACCACTTCATCGGCAGCCTGGCGGTGGCAGACCTC
CTGGGGAGTGTCATTTTTGTCTACAGCTTCATTGACTTCCACGTGTTCCACCGCAA
AGATAGCCGCAACGTGTTTCTGTTCAAACTGGGTGGGGTCACGGCCTCCTTCACT
GCCTCCGTGGGCAGCCTGTTCCTCACAGCCATCGACAGGTACATATCCATTCACA
GGCCCCTGGCCTATAAGAGGATTGTCACCAGGCCCAAGGCCGTGGTGGCGTTTT
GCCTGATGTGGACCATAGCCATTGTGATCGCCGTGCTGCCTCTCCTGGGCTGGAA
CTGCGAGAAACTGCAATCTGTTTGCTCAGACATTTTCCCACACATTGATGAAACC
TACCTGATGTTCTGGATCGGGGTCACCAGCGTACTGCTTCTGTTCATCGTGTATG
CGTACATGTATATTCTCTGGAAGGCTCACAGCCACGCCGTCCGCATGATTCAGCG
TGGCACCCAGAAGAGCATCATCATCCACACGTCTGAGGATGGGAAGGTACAGGT
GACCCGGCCAGACCAAGCCCGCATGGACATTAGGTTAGCCAAGACCCTGGTCCT
GATCCTGGTGGTGTTGATCATCTGCTGGGGCCCTCTGCTTGCAATCATGGTGTAT
GATGTCTTTGGGAAGATGAACAAGCTCATTAAGACGGTGTTTGCATTCTGCAGTA TGCTCTGCCTGCTGAACTCCACCGTGAACCCCATCATCTATGCTCTGAGGAGTAA GGACCTGCGACACGCTTTCCGGAGCATGTTTCCCTCTTGTGAAGGCTAG
SEO ID NO:2
MKSILDGLADTTFRTITTDLLYVGSNDIQYEDIKGDMASKLGYFPQKFPLTSFRGSPFQ
EKMTAGDNPQLVP ADQVNITEFYNKSLSSFKENEENIQCGENFMDIECFMVLNPSQQ
LAIAVLSLTLGTFTVLENLLVLCVILHSRSLRCRPSYHFIGSLAVADLLGSVIFVYSFIDF
HVFHRKDSRNVFLFKLGGVTASFTASVGSLFLTAIDRYISIHRPLAYKRIVTRPKAVVA
FCLMWTIAIVIAVLPLLGWNCEKLQSVCSDIFPHIDETYLMFWIGVTSVLLLFIVYAYM
YILWKAHSHAVRMIQRGTQKSIIIHTSEDGKVQVTRPDQARMDIRLAKTLVLILVVLII
CWGPLLAIMVYDVFGKMNKLIKTVFAFCSMLCLLNSTVNPIIYALRSKDLRHAFRSM
FPSCEG*
Example 615: Acute Feeding Study
[0613] Male, Sprague-Dawley rats (90-120 g) served as subjects for these studies. Rats were fasted for a period of 16 hrs (water was always available). After the fasting period, test compounds were administered either intraperitoneally (ip) or orally (po). Immediately following compound administration, the rats were returned to their home cage. Following 30 min after compound administration, the rats were removed from their home cages and placed individually into clean cages with a pre-measured amount of food. Food weights were obtained (to the nearest 0.1 g) at various time points. Food consumption was monitored for a period of up to 2 hrs (i.e., 2.5 hr after test compound administration).
[0614] Figure 2 is a bar graph showing the food intake in fasted rats 1 and 2 hours after being administered either 1, 3, or 10 mg/kg doses of Compound I. * Indicates p<0.05 as compared to the vehicle-treated controls. ** Indicates p<0.01 as compared to the vehicle- treated controls. Figure 3 is bar graph showing the time course food intake in fasted rats after being administered 1 mg/kg of Compound I. * Indicates p<0.05 as compared to the vehicle- treated controls. ** Indicates p<0.01 as compared to the vehicle-treated controls. Figure 4 is a bar graph showing cumulative food consumption at several points in time after the rats had been dosed with 10 mg/kg of Compound I. * Indicates p<0.05 as compared to the vehicle- treated controls. As shown by Figures 2-4, Compound I suppresses the cumulative food intake in fasted rats. Figure 2 also shows that suppression of food take is dose-dependent.
Example 616: Tail Flick Study
[0615] Male, NSA mice (15-20 g) served as subjects for these studies. Baseline nociceptive thresholds were assessed using the warm water tail flick test. Briefly, the distal 1/3 to 1A of the tail was 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 was recorded (i.e., tail flick latency). Mice were then injected ip with either vehicle or with various doses of the CBl agonist CP 55,940 and tail flick latencies were recorded for a period of up to 3 hr. A maximum latency of 10 sec was employed in order to prevent tissue damage. In order to determine if a CBl inverse agonists could block the antinociceptive actions of CP 55,940, mice were pretreated with either vehicle or with a test compound 30 min prior to CP55,940. CP55,940 (1 mg/kg) was administered subcutaneously, and Compound I was administered intraperitoneally. Tail flick latencies were then obtained at various time points for a period of up to 2 hr. The vehicle for both compounds was 1:1:18 cremphor:ethanol:saline.
[0616] Figure 5 A is a line graph showing the attenuation of CBl agonist-mediated effects after administration of CP 55,940 (0.3 and 1.0 mg/kg). Figure 5B is a line graph showing the attenuation of CBl agonist-mediated effects after administration of Compound I alone or in combination with CP55,940. As indicated by Figures 5A and 5B, Compound I attenuates the antinociceptive actions of CP55,940.
Example 617: Hypothermia Study
[0617] Male, NSA mice (15-20 g) served as subjects for these studies. In order to determine if the test compound could block hypothermia elicited by CP 55,940 (1 mg/kg, ip), mice were pretreated with either vehicle or with test compound 30 min prior to CP55,940. Core body temperatures were then obtained at various time points following CP 55,940 administration. Core body temperature (to the nearest 0.10C) was obtained by rectal probe.
[0618] Figure 6 is a bar graph showing the body temperature of the rats at several points in time after the rats had been dosed with various doses of CP 55,950 or CP55,950 and Compound I. As shown by Figure 6, Compound I attenuates CP 55,940-induced hypothermia. In addition, the attenuation of the CP55,940-induced hypothermia was dose- dependent.
Example 618: Chronic Feeding Study
[0619] Male, obese Zucker rats (400-500 g) served as subjects for these studies. Rats were housed individually and had access to food and water ad libitum. Rats were allowed to acclimate to the vivarium for a period of 3 days, during which body weight and consumption of food and water was monitored. Rats were weighed daily at 1500 hr and then injected with either vehicle or with various doses of the test compound. Daily food and water intakes were also monitored. Food and water bottles were weighed at the time body weights were recorded (i.e., 1350 hr). Vehicle or compound was administered daily for a period of up to 15 days.
[0620] Figure 9A in a line graph showing the effects of Compound II (1 and 3 mg/kg/day) on body weight Figure 9B is a line graph showing the effects of Compound II (1 and 3 mg/kg/day) on food intake and water intake. Figure 9C line graph showing the effects of Compound II (10 mg/kg/day) on body weight. Figure 9D is a line graph showing the effects of Compound II (10 mg/kg/day) on food intake and water intake. As shown by Figures 9A-9D, Compound II attenuated the food and water intake of the rats. Moreover, the attenuation of the food and water intake was dose-dependent.
Example 619: Novel Object Recognition Study
[0621] Subjects: Subjects were male, C57 BK/6 mice purchased from Harlan Laboratories, weighing 15-2Og upon arrival. Animals wqre housed 8 per cage with food and water available ad libidum. Animals were housed on a 12 hr light cycle (lights on 6 am) for 4-7 days prior to behavioral testing.
[0622] Equipment: Novel object recognition (NOR) was 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 was covered with a piece of plastic lined bench top paper. There were 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 were yellow, ceramic, 12-sided ramekins measuring 4 cm high x 7 cm diameter. "B" objects were 8 X 8 x 4 cm stainless steel, 4-sided ramekins.
[0623] Procedure: At the beginning of each test day, animals were placed in groups of 6 into clean cages. Testing was conducted in three phases: acclimation, sample and test. For acclimation, each group of six mice was placed collectively into the NOR chamber and allowed to explore freely for 30 min. After acclimation animals were 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 was placed, one at a time into the NOR chamber, into which two identical objects had been placed ("A" or "B" objects described above). Objects were placed on diagonal corners of the long axis of the arena approximately 5 cm from the walls, while subjects were placed into one of the neutral corners (alternating across subjects). Each mouse was allowed to explore the chamber and the objects for 3 min., and the time spent exploring at each position was recorded. Directly sniffing or touching the object was recorded as exploration. After 3 min., each mouse was 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 were placed into the chamber in the same positions used, during the sample phase, and each mouse was allowed 3 min to explore. The test sessions were 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 was placed were counterbalanced across subjects. Prior to each trial (acclimation, sample and test), all equipment was wiped with a Clorox wipe and bench paper (cut to fit) was placed in the bottom of the chamber. The procedure is shown below in Scheme 9.
[0624] Measures: In addition to time spent exploring each object (TN = time spent exploring novel object, TF = time spent exploring familiar object), two measures were 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 9
Figure imgf000236_0001
[0625] Figures 1OA and 1OC are bar graphs showing the exploration ratio at 1 and 2 hours after the mice had been dosed with the vehicle, CP 55,940 (0.3 mg/kg, ip), or SR141716A (1 mg/kg, ip). Figures 1OB and 1OD are bar graphs showing the discrimination index at 1 and 2 hours after the mice had been dosed with the vehicle, CP 55,940 (0.3 mg/kg, ip), or SR141716A (1 mg/kg, ip). Figure HA is a bar graph showing the exploration ratio 2 hours after the mice had been dosed with Compound II (3 mg/kg, ip). Figure HB is a bar graph showing the discrimination index 2 hours after the mice had been dosed with Compound II (3 mg/kg, ip).
[0626] As shown by Figures 10A-D and Figures HA-B, mice treated with SR141716A and Compound II showed a preference for the novel object (indicating the mice recognized the familiar object) up to two hours after being dosed with the test compound. Mice treated with the vehicle or CP 55,940 showed a preference for the novel object after 1 hour of being dosed with the test compound but then returned back to baseline exploration rates after 2 hours.
Example 620: Radial Arm Maze Study
[0627] Subjects: Subjects for the radial arm maze experiments were male, Sprague-Dawley rats purchased Charles Rivers Laboratories, weighing 225-250 g upon arrival, housed two per cage. All subjects had free access to food and water available for the duration of the study. Animals were housed on a 12 hr light cycle (lights on 7 am), and were acclimated to vivarium conditions for a minimum of two days prior to behavioral training. AU experiments were conducted in accordance with NIH Guidelines for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Use Committee at ACADIA Pharmaceuticals, Inc.
[0628] Radial Arm Maze Procedure: Radial arm maze (RAM) testing was conducted in a watertight maze (61.0 cm high) made of black ABS plastic, consisting of a central, round chamber (57.1 cm in diameter) with 8 (38.1 cm X 16.6 cm) equally spaced arms radiating from the center. The testing room had salient environmental cues that remained constant throughout testing, including a door, a table, a shelving unit, a solid black panel one wall, a black and white striped panel on the opposite wall, and the experimenter seated behind the start arm. Prior to each session, escape platforms were placed in the ends of 6 arms. Escape platforms were made of black ABS plastic (10.1 cm X 15.2 cm) covered with Velcro fitted 16 cm from the top of the maze. Each day the maze was filled with water (25°C) until the platforms were hidden with 1 cm of water covering the platforms. Additionally, non-toxic black paint was dissolved in the water to help visually obscure the platforms and ensure animals could not depend on visual cues to solve the task. For each subject, reference arms (arms without platforms) remained constant across training and testing. During a trial, a subject was released from the start arm, facing the center, and allowed 3 min to locate a platform. If the maximum time elapsed, the animal was guided to the nearest platform. Once a platform was found, animals remained on it for 15 sec before being removed from the maze and placed in a warmed holding tub for 30 sec. During the interval, the chosen platform was removed from the maze. The animal was then returned to the maze for another trial. This continued until all platforms were located. Training was conducted 5 days per week for 10 days. After training, animals began the test phase. During testing, animals received multiple test sessions. In order to ensure adequate time for drug clearance between treatments, subjects received only one test compound and one vehicle treatment, per week. In all other respects, test sessions were conducted using the same method described for training.
[0629] Figure 12 is a bar graph showing percentage of novel recognition of a familiar object 2 hours after the mice had been dosed with 1, 3, or 10 mg/kg of Compound II. Figure 13 is a line graph showing the working memory errors of the mice after being dosed with the vehicle, tacrine (0.3 mg/kg), or Compound II (3 mg/kg).
[0630] As shown by Figures 12 and 13, mice treated with Compound II showed a preference for the novel object (indicating the mice recognized the familiar object) up to two hours after being dosed with the test compound.
Example 621 : Rotation Study
[0631] Subjects: Subjects were male, Sprague-Dawley rats purchased from Harlan Laboratories, weighing 250-275 g upon arrival. Prior to surgery animals were housed two per cage. All subjects had free access to food and water available for the duration of the study. Animals were housed on a 12 hr light cycle (lights on 6 am), and were acclimated to vivarium conditions for a minimum of one week prior to surgery. All experiments were conducted in accordance with NIH Guidelines for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Use Committee at ACADIA Pharmaceuticals, Inc.
[0632] Surgery. One week after arrival, subjects underwent 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 were administered desipramine (20 mg/kg ip) approximately 20 min prior to surgery. Surgery was conducted under ketamine (80 mg/kg ip) and xylazine (12 mg/kg ip) anesthesia. Animals were placed in the stereotaxic instrument with the incisor bar at -3.2 mm and a hole was 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 was lowered to -8.2 mm from bregma. 8 μg of 6-hydroxy-dopamine in 4 μl of saline with 0.2% ascorbic acid was infused over 5 min, and 1 min was allowed for diffusion before the syringe was removed and the incision closed. Animals were given a minimum of 15 days after surgery before any behavioral assessment.
[0633] Rotational Behavior. All animals were assessed for rotational behavior in rotameters purchased from San Diego Instruments, Inc. For each behavioral session, subjects were placed in the rotometers and allowed thirty minutes for acclimation. After 30 min., subjects were injected with either the dopamine agonist apomorphine (0.05, 0.16 or 0.5 nig/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 received combinations of the two treatments, Compound II was injected 30 minutes prior to apomorphine. After treatment, rotations were measured for 60 min. Subjects were then removed from the rotometers and returned to their home cages. All animals received 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.
[0634] Figure 14 is a line showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.05, 0.16, and 0.5 mg/kg). Figure 15 is a line showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.05 mg/kg), Compound II (3.0 mg/kg), or apomorphine (0.05 mg/kg) and Compound II (3.0 mg/kg). Figure 16 is a line showing the contralateral rotations over time of the mice after being dosed with apomorphine (0.16 mg/kg), Compound II (3.0 mg/kg), or apomorphine (0.16 mg/kg) and Compound II (3.0 mg/kg).
[0635] As shown by Figure 14, apomorphine dose-dependently elicits contralateral rotations in rats with unilateral 6-OH dopamine lesions. Figures 15 and 16 show that Compound II augments dopaminergic functions.
[0636] 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;
[0637] The following references are incorporated by reference herein in their entirety:
1 , Le Foil B, Goldberg SR. Cannabinoid CB 1 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.
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001

Claims

WHAT IS CLAIMED IS:
1. A compound of formula (I) :
Figure imgf000315_0001
as a single isomer, a mixture of isomers, a racemic mixture of isomers, pharmaceutically acceptable salt, a solvate, metabolite or polymorph thereof, wherein: ,
X is selected from the group consisting of O, S, S=O, SO2, NR1, NC=N, NCC=Z)Ri, NC(=Z)NRiaRlb, CRlaRlb, C=O, C=CRlaRlb, and SiRlaRlb;
Y is -N(R2) — or -C(R1R2) — ; the symbol :r^: represents a single or double bond, where when ==-= is a double bond, R2 is absent;
A is selected from the group consisting of C3-C]2alkyl, C4-Ci2alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, (cycloalkyl)alkyl, (cycloalkenyl)alkyl, (cycloalkynyl)alkyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, halogen, -NRi JR1 b, -N=CRj aRib, sulfenyl, sulfinyl, sulfonyl, and -(CH2)0-4-C(=Z)-ORi, wherein any member of said group can be substituted or unsubstituted; provided that A cannot be a substituted or unsubstituted piperazine;
B, C, D, E, F, G and I are separately selected from the group consisting of 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)R], -C(=Z)ORi, -CC=Z)NRiJR-Ib, -CC=Z)N(R1)NR13Rn,, -CC=Z)N(R1)N(R1)CC=Z)R1, - C(RO=NR1, -NRiaRib, -N=CRiaRib, -N(Rj)-Q=Z)Ri, -N(Ri)-C(=Z)NRlaRlb, - S(O)NRi8RiI5, -S(O)2NR1 aRib, -N(R1)-S(=0)R1, -N(R,)-S(=O)2Ri, -OR1, -SR1, and -OCC=Z)R1, wherein any member of said group can be substituted or unsubstituted except for hydrogen; H is selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, halogen, hydroxyl, nitro, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, -CN, -C(=Z)R,, -C(=Z)ORi, -C(=Z)NRlaRib, -Q=Z)N(R1 )NRIaR,b, -CC=Z)N(Ri)N(R1)CC=Z)R1, -C(Ri)=NRi, -NRlaR,b, -N=CRIaRIb, -N(R,)-C(=Z)R,, -N(R,)-C(=Z)NRiβRib, -S(0)NRlaRlb, -S(O)2NRlaRlb, -N(R,)-S(=O)Ri, -N(R])-S(=O)2Rl5 -OR], -SRi, and -OC(=Z)Ri, wherein any member of said group can be substituted or unsubstituted; with the proviso that H cannot be selected from the group consisting of -CF3,
phenyl, -OS(O)2-CF3, methyl, -CN, halogen, and
Figure imgf000316_0001
when A is a substituted or unsubstituted heteroalicyclyl containing at least one nitrogen or -NRiaRib; with the proviso that H cannot be halogen when A is substituted or unsubstituted aryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroaryl, halogen, and substituted or unsubstituted sulfenyl; X is -NRi1 wherein Ri is hydrogen; and Y is -N(R2) === , wherein = is a double bond and R2 is absent;
Z is O or S;
Ri1 Rja and Rib are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, (CH2)O-7-OR3, -(CH2)O-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 unsubstituted except for hydrogen; or R]3 and Ri b 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, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted except for hydrogen; R3, R3a, and R3b are each independently selected from the group consisting of: 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; with the proviso when X is O or -NRi, wherein Ri is methyl and Y is - N(R2) = , wherein = is a double bond and R2 is absent then H cannot be - Q=Z)OR1 , wherein Rj is hydrogen, methyl, or ethyl; and with the proviso that when A is halogen, Y is -N(R2) =, wherein =^= is a double bond and R2 is absent, and X is S then F cannot be -S(O)2NRi3RIb, wherein Riaand Rib are both hydrogen.
2. The compound of Claim I3 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 Claim 1, wherein Rj3 and R^ form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms and 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, sulfmyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino.
5. The compound of Claim 1, wherein Ria and Rib form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from the group consisting of:
Figure imgf000317_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, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, amino and protected amino.
6. The compound of Claim 5, wherein Ria and Rib form an unsubstituted or substituted heteroalicyclyl having 2 to 9 carbon atoms selected from the group consisting of:
Figure imgf000318_0001
7. The compound of Claim 1, wherein X is S, SO, or SO2.
8. The compound of Claim 1, wherein:
H is selected from the group consisting of aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, halogen, -C(=Z)Ri, -C(=Z)OR,, -C(=Z)NRlaRlb, -C(=Z)N(R,)NRlaRlb,
-C(=Z)N(Ri)N(R,)C(=Z)R,, -C(RO=NR1, -NRlaRlb, -N=CRlaRIb, -N(Ri)-CC=Z)R1, -N(R,)-C(=Z)NRlaRIb, -S(O)NRlaRlb, -S(O)2NR1 aRlb, -N(Ri)-S(=O)Ri, -N(Ri)-S(=O)2Ri, and -OC(=Z)Ri, wherein any member of said group can be substituted or unsubstituted.
9. The compound of Claim 1 , wherein:
H is selected from the group consisting of cycloalkyl, cycloalkenyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, hydroxyl, sulfenyl, sulfinyl, sulfonyl, haloalkoxy, -C(=Z)ORj, -C(=Z)N(Ri)NRiaRib, -C(=Z)N(Ri)N(Ri)C(=*Z)R,, -C(Ri)=NR1, -NR,aR,b, - N=CRlaRlb, -S(O)NRlaRlb, -N(Ri)-S(K))Ri, -N(R))-S(=O)2R,, and -0C(=Z)Ri, wherein any member of said group can be substituted or unsubstituted.
10. The compound of Claim 1, wherein H is selected from the group consisting of cycloalkyl, aryl, heteroaryl, and heteroalicyclyl, wherein any member of said group can be substituted or unsubstituted.
11. The compound of Claim 10, wherein the unsubstituted or substituted heteroaryl is selected from the group consisting of:
Figure imgf000319_0001
12. The compound of Claim 10, wherein the aryl is an optionally substituted phenyl.
13. The compound of Claim 12, wherein the optionally substituted phenyl is substituted with a Cj-4 alkyl.
14. The compound of Claim 1, wherein H is -C(=Z)NRiaRib.
15. The compound of Claim 14, wherein R1 a is selected from the group consisting of alkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and - (CH2)O-7-NR3 aR3b, wherein any member of said group can be substituted or unsubstituted.
16. The compound of Claim 14, wherein R1 a is selected from the group consisting of alkyl, alkoxy, aryl, aralkyl, heteroaryl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted.
17. The compound of Claims 14, wherein Ri a is an optionally substituted heteroaryl or heteroaralkyl.
18. The compound of any of Claims 14-17, wherein R^ is hydrogen or methyl.
19. The compound of Claim 17, wherein the optionally substituted heteroaryl or heteroaralkyl is selected from the group consisting of:
Figure imgf000319_0002
wherein Q is oxygen or sulfur.
20. The compound of Claim 19, wherein the optionally substituted heteroaralkyl is
Figure imgf000320_0001
21. The compound of any of Claims 19-20, wherein n is 1 or 2.
22. The compound of Claim 1, wherein H is -C(=Z)Ri or -C(=Z)OR[.
23. The compound of Claim 22, wherein H is -C(=Z)Ri and Ri is selected from the group consisting of alkyl, cycloalkyl, aralkyl, halogen.
24. The compound of Claim 22, wherein H is -C(=Z)ORi and R1 is alkyl or aralkyl.
25. The compound of Claim 1, wherein H is -C(=Z)N(Ri)N(Ri)C(=Z)Ri or
-N(R0-C(=Z)NRlaRlb.
26. The compound of Claim 25, wherein -C(=Z)N(R1)N(Ri)C(=Z)R1 is
Figure imgf000320_0002
, wherein n is 0 or 1.
27. The compound of Claim 25, wherein H is -N(Ri)-C(=Z)NR]aRib and Ri is hydrogen and Ri a is alkyl or aralkyl.
28. The compound of Claim 27, wherein Rn, is hydrogen.
29. The compound of Claim 1, wherein H is selected from the group consisting of -C(RO=NRi, -N(R,)-C(=Z)Ri, and -OC(=Z)Ri.
30. The compound of Claim 29, wherein H is -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 the group consisting of alkyl, aryl, and aralkyl.
31. The compound of Claim 1 , wherein H is -N(Ri)-SC=O)R1 or -N(Ri)-S(=O)2Ri .
32. The compound of Claim 31, wherein is -N(Rj)-S(O)Ri or -N(Ri)-SC=O)2Ri and Ri is hydrogen, aralkyl, or heteroaryl.
33. The compound of Claim 1, wherein H is -S(O)NRiaRib or -S(O)2NRi aRib.
34. The compound of Claim 33, wherein H is -S(O)NRiaRib or -S(O)2NRiaRib and Rla is selected from the group consisting of alkyl, aryl, aralkyl, heteroaryl, and heteroalicyclyl.
35. The compound of any of Claims 33-34, wherein R^ is hydrogen.
36. The compound of Claim 1, wherein if H is -S(O)NRlaRlb, -S(O)2NRiaRlb, -C(=Z)NRlaR!b or -C(=Z)N(Ri)NRlaR1b then Rj1 Rjaand Rjb'are each independently selected from the group consisting of:
wherein: n is an integer selected from the group consisting of 0, 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(ILt)-, 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, R63, Rόb, R6C, and R6d 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, R6a, Rβb, R6C5 and R6d 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, R6b, R60, and R61].
37. The compound of Claim 36, wherein H is -C(=Z)NRlaRib.
38. The compound of any of Claims 36-37, wherein n is 0, 1, or 2.
39. The compound of Claim 1, wherein R1, Rla, R2a, R2, R3, R3a, and R3b are each independently selected from the group consisting of aryl, heteroaryl, heteroalicyclyl, aralkyl, heteraralkyl, or (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.
40. The compound of Claim 1 , 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, 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.
41. The compound of Claim 40, 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.
42. The compound of Claim 1, wherein:
X is selected from the group consisting of S, S=O, and SO2;
Y is -N(R2)= or -C(R1R2)^; 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 C3-Ci2alkyl, C4-C]2alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, aralkyl, and heteroaralkyl, wherein any member of said group can be substituted or unsubstituted;
B, C, D, E, F, G and I are separately selected from the group consisting of 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)R], -C(=Z)0Ri, -C(=Z)NRiaRlb, -C(=Z)N(Rj)NRIaRib, -C(=Z)N(Ri)N(Ri)C(=Z)Rl 5 - C(Ri)=NR1, -NRRlb, -N=CR,aRIb, -N(R,)-C(=Z)Ri, -N(R,)-C(=Z)NRlaRlb, - S(O)NRlaRlb, -S(O)2NRlaRib, -N(R,)-S(=O)R,, -N(RO-SC=O)2R1, -OR1, -SR1, and -OC(=Z)Rii wherein any member of said group can be substituted or unsubstituted except for hydrogen;
H is selected from the group consisting of -C(=Z)NRiaRit>, -C(=Z)N(R0NRlaRlb, -C(=Z)N(Ri)N(Ri)C(=Z)Ri, and -C(Rj)=NR1, wherein any member of said group can be substituted or unsubstituted;
Z is O or S;
Ri1 R1 a and Rj b are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl, (CH2)O-7-OR3, -(CH2)O-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 unsubstituted except for hydrogen; or RJ a and R] b 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, 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
R3, R3a, and R3b are each independently selected from the group consisting of: 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.
43. The compound of Claim 42, wherein Z is O.
44. The compound of any of Claims 42-43, wherein A is selected from the group consisting of C3-Ci2alkyl, C4-Ci2alkyl, cycloalkyl, aryl, and heteroaryl, wherein any member of said group can be substituted or unsubstituted.
45. The compound of Claim 1, wherein: A is selected from the group consisting of C3-Ci2alkyl, C4-Ci2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)0-4-C(=Z)- OR1; and
X is S.
46. The compound of Claim 45, wherein:
A is selected from the group consisting of C3-Ci2alkyl, C4-Cj2alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)0-4-C(=Z)- OR1;
X is S; and
Y is -N(R2) =^ wherein the symbol =^= represents a double bond and R2 does not exist.
47. The compound of Claim 46, wherein:
A is selected from the group consisting of Cs-C^alkyl, C4-C12alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclyl, halogen, -NRiaRib, and -(CH2)0-4-C(=Z)- OR1;
X is S;
Y is -N(R2) ^= wherein the symbol =^= represents a double bond and R2 does not exist; and
H is -C(=Z)NRlaRlb.
48. The compound of Claim 47, wherein:
A is selected from the group consisting of C3-C12alkyl, C4-Ci2alkyl, halogen, and -(CH2)o-4-C(=Z)-ORi; X is S;
Y is -N(R2) === wherein the symbol =^= represents a double bond and R2 does not exist; and
H is -C(=Z)NRlaRlb.
49. The compound of Claim 47, wherein: A is an aryl or a heteroaryl group;
X is S;
Y is -N(R2) =^= wherein the symbol =^= represents a double bond and R2 does not exist; and H is -C(=Z)NRiaRib.
50. The compound of Claim 47, wherein:
A is a cycloalkyl, a heteroalicyclyl, or -NRuRib group; X is S;
Y is -N(R2) ="^ wherein the symbol =^= represents a double bond and R2 does not exist; and
H is -C(=Z)NR_aRib.
51. The compound of any of Claims 47-50, wherein: X is S;
Y is -N(R2)=^ wherein the symbol =^: represents a double bond and R2 does not exist; and
H is -C(=Z)NRiaRib, wherein R]3 is selected from the group consisiting of alkyl, alkoxy, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heteroalicyclyl, (heteroalicyclyl)alkyl and -(CH2)0-7-NR3aR3b, wherein any member of said group can be substituted or unsubstituted.
52. The compound of any of Claims 47-50, wherein: X is S;
Y is -N(R2) == wherein the symbol ^=-= represents a double bond and R2 does not exist; and
H is -C(=Z)NRlaRib, wherein Rlais an optionally substituted alkyl, alkoxy, or -(CH2)0-7-NR3aR3b.
53. The compound of Claim 52, wherein the alkyl is a Cj-6 alkyl.
54. The compound of Claim 52, wherein the alkoxy is a C1-6 alkoxy.
55. The compound of any of Claims 47-50, wherein: X is S;
Y is -N(R2) =^= wherein the symbol == represents a double bond and R2 does not exist; and
H is -C(=Z)NR]aRib, wherein Ri a is an optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl.
56. The compound of Claim 55, wherein the optionally substituted cycloalkyl, cycloalkenyl, or cycloalkynyl is selected from the group consisting of:
Figure imgf000327_0001
57. The compound of Claim 56, wherein n is 1 or 2.
58. The compound of any of Claims 47-50, wherein: X is S;
Y is -N(R2)=^ wherein the symbol === represents a double bond and R2 does not exist; and
H is -C(=Z)NRlaRib, wherein Riais an optionally substituted aryl or aralkyl.
59. The compound of Claim 58, wherein the optionally substituted aryl or aralkyl s selected from the group consisting of:
Figure imgf000327_0002
wherein Q is -N(R4)-, oxygen or sulfur; and R4 is hydrogen or Ci-4alkyL
60. The compound of Claim 59, wherein n is 1 or 2.
61. The compound of any of Claims 47-50, wherein: X is S;
Y is -N(R2) = wherein the symbol ^= represents a double bond and R2 does not exist; and
H is -C(=Z)NRlaR1b, wherein Rla is an optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl.
62. The compound of Claim 61, wherein the optionally substituted heteroalicyclyl or (heteroalicyclyl)alkyl is selected from the group consisting of:
Figure imgf000328_0001
63. The compound of Claim 62, wherein n is 1 or 2.
64. The compound of any of Claims 47-50, wherein: X is S;
Y is — N(R2) ^^ wherein the symbol ^^^ represents a double bond and R2 does not exist; and
H is -C(=Z)NRiaRib) wherein Rla is an optionally substituted heteroaryl or heteroaralkyl.
65. The compound of Claim 64, wherein the optionally substituted heteroaralkyl the group consisting of:
Figure imgf000328_0002
wherein Q is oxygen or sulfur.
66. The compound of Claim 65, wherein the optionally substituted heteroaralkyl is
Figure imgf000329_0001
67. The compound of any of Claims 65-66, wherein n is 1 or 2.
68. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000330_0001
69. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000331_0001
Figure imgf000332_0001
Figure imgf000333_0001
70. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000334_0001
Figure imgf000335_0001
Figure imgf000335_0002
Figure imgf000336_0001
Figure imgf000336_0002
Figure imgf000336_0003
Figure imgf000336_0004
Figure imgf000336_0005
71. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
72. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000340_0001
73. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Figure imgf000344_0001
74. The compound of Claim I3 wherein the compound is selected from the group consisting of:
Figure imgf000345_0001
Figure imgf000346_0001
Figure imgf000347_0001
Figure imgf000347_0002
Figure imgf000348_0001
Figure imgf000348_0002
Figure imgf000348_0003
Figure imgf000348_0004
Figure imgf000349_0001
Figure imgf000349_0002
Figure imgf000350_0001
Figure imgf000350_0002
Figure imgf000351_0001
Figure imgf000351_0002
Figure imgf000351_0003
Figure imgf000351_0004
Figure imgf000352_0001
Figure imgf000352_0002
Figure imgf000352_0003
Figure imgf000353_0001
Figure imgf000353_0002
Figure imgf000353_0003
Figure imgf000354_0001
Figure imgf000354_0002
75. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000355_0001
Figure imgf000355_0002
76. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000356_0001
Figure imgf000357_0001
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
Figure imgf000361_0001
Figure imgf000362_0001
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Figure imgf000366_0001
Figure imgf000367_0001
77. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000367_0002
Figure imgf000368_0001
Figure imgf000369_0001
Figure imgf000369_0002
Figure imgf000369_0003
Figure imgf000370_0001
Figure imgf000370_0002
Figure imgf000370_0003
78. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000371_0001
79. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000372_0001
Figure imgf000373_0001
Figure imgf000374_0001
80. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000375_0001
81. The compound of Claim I5 wherein the compound is selected from the group consisting of:
Figure imgf000375_0002
82. The compound of Claim I5 wherein the compound is selected from the group consisting of:
Figure imgf000375_0003
Figure imgf000376_0001
83. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000377_0001
84. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000377_0002
85. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000378_0001
86. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000378_0002
Figure imgf000379_0001
Figure imgf000379_0002
Figure imgf000379_0003
Figure imgf000379_0005
Figure imgf000379_0004
87. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000379_0006
Figure imgf000380_0001
88. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000381_0001
Figure imgf000382_0001
Figure imgf000383_0001
Figure imgf000384_0001
Figure imgf000385_0001
Figure imgf000386_0002
Figure imgf000386_0001
Figure imgf000386_0003
Figure imgf000387_0001
Figure imgf000388_0001
89. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000389_0001
Figure imgf000390_0001
Figure imgf000391_0001
90. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000392_0001
Figure imgf000392_0002
Figure imgf000392_0003
Figure imgf000392_0004
Figure imgf000393_0001
Figure imgf000394_0001
Figure imgf000394_0003
Figure imgf000394_0002
91. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000395_0001
Figure imgf000396_0001
Figure imgf000396_0002
Figure imgf000396_0004
Figure imgf000396_0003
Figure imgf000397_0001
92. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000398_0001
Figure imgf000398_0002
Figure imgf000399_0001
Figure imgf000400_0001
Figure imgf000401_0001
93. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000402_0001
Figure imgf000403_0001
Figure imgf000404_0001
94. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000404_0002
Figure imgf000405_0001
95. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000405_0002
96. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000405_0003
Figure imgf000406_0001
Figure imgf000407_0001
97. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000408_0001
Figure imgf000409_0001
Figure imgf000410_0001
98. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000411_0001
99. The compound of Claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000413_0001
Figure imgf000413_0002
Figure imgf000413_0004
Figure imgf000413_0003
Figure imgf000414_0001
100. A pharmaceutical composition, comprising a therapeutically effective amount of a compound of Claim 1 and a pharmaceutically acceptable carrier, diluent, or excipient.
101. A method of ameliorating or preventing 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, 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 the subject a therapeutically effective amount of a compound of Claim 1.
102. The method of Claim 101, wherein the therapeutically effective amount of a compound of Claim 1 is in a sufficient amount to ameliorate or prevent said disease or condition by binding to a cannabinoid receptor.
103. The method of Claim 101, further comprising identifying a subject in need of ameliorating or preventing said disease or condition.
104. The method of Claim 101, 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.
105. The method of Claim 101, wherein the disease or condition is selected from the group consisting of obesity, metabolic syndrome, an appetite disorder, cachexia, high cholesterol, hyperlipidemia, and diabetes.
106. The method of Claim 101, wherein the disease or condition is selected from the group consisting of emesis, nausea, a gastric ulcer, diarrhea, and intestinal disorders.
107. The method of Claim 101, wherein said inflammatory disease is selected from the group consisting of rheumatoid arthritis, asthma, and psoriasis.
108. The method of Claim 101, wherein the disease or condition is selected from the group consisting of hemorrhagic shock, septic shock, cirrhosis, atherosclerosis, and a cardiovascular disorder.
109. The method of Claim 101, wherein the disease or condition is selected from the group consisting of infertility and premature abortion.
110. The method of Claim 101, wherein the disease or condition is selected from the group consisting of glaucoma, uveitis, retinopathy, dry eye, and macular degeneration.
111. The method of Claim 101, wherein the disease or condition is selected from the group consisting of osteoporosis and ostepenia.
112. The method of Claim 101, wherein the disease or condition is selected from the group consisting of asthma and pleurisy.
113. A method of Claim 101, wherein the disease or condition is cancer.
114. A method for treating or preventing a disease or condition in which it would be beneficial to modulate the activity of a CBl receptor comprising administering a therapeutically effective amount of a compound of Claim 1.
115. A method of ameliorating drug addition or alcohol addiction comprising administering to a subject a pharmaceutically effective amount of a compound of Claim 1.
116. The method of Claim 115, further comprising identifying a subject in need of ameliorating a drug addiction or alcohol addiction.
117. A method of ameliorating obesity comprising administering to a subject a pharmaceutically effective amount of a compound of Claim 1.
118. The method of Claim 117, further comprising identifying a subject in need of amerliorating obesity.
119. A method of ameliorating impaired cognition or a memory disorder comprising administering to a subject a pharmaceutically effective amount of a compound of Claim 1.
120. The method of Claim 119, further comprising identifying a subject in need of amerliorating impaired cognition.
121. A method of improving cognition or memory in a subject comprising administering to a subject a pharmaceutically effective amount of a compound of Claim 1.
122. A method of preventing or allevaiting inflammation due to an inflammatory disease comprising administering to a subject a pharmaceutically effective amount of a compound of Claim 1.
123. The method of Claim 122, wherein the inflammatory disease is selected from the group consisting of rheumatoid arthritis, asthma, and psoriasis.
.
124. The method of Claim 122, further comprising identifying a subject in need of amerliorating an inflammatory disease.
125. 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 Claim 1.
126. The method of Claim 125, wherein the cannabinoid receptor is a CBl receptor.
127. A method of modulating or specifically inverse agonizing or antagonizing a cannabinoid receptor comprising contacting a cannabinoid receptor with a compound of Claim 1.
128. The method of Claim 127, wherein the cannabinoid receptor is a CBl receptor.
129. 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 Claim 1 ; 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.
130. The method of Claim 129, wherein the cannabinoid receptor is a CBl receptor. ,
131. The method of Claim 129, wherein said cannabinoid receptor consists essentially of SEQ ID NO: 2.
132. The method of Claim 129, wherein said cannabinoid receptor has at least 90% amino acid identity to SEQ ID NO: 2.
133. The method of Claim 129, wherein said cannabinoid receptor has at least 85% amino acid identity to SEQ ID NO: 2.
134. The method of Claim 129, wherein said cannabinoid receptor has at least 70% amino acid identity to SEQ ID NO: 2.
135. A method of identifying a compound which is an agonist, inverse agonist, or antagonist of a cannabinoid receptor comprising: culturing cells that express said a cannabinoid receptor; incubating the cells or a component extracted from the cells with at least one test compound of Claim 1 ; 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.
136. The method of Claim 135, wherein the cannabinoid receptor is a CBl receptor.
137. The method of Claim 135, wherein said cannabinoid receptor consists essentially of SEQ ID NO: 2.
138. The method of Claim 135, wherein said cannabinoid receptor has at least 90% amino acid identity to SEQ ID NO: 2.
139. The method of Claim 135, wherein said cannabinoid receptor has at least 85% amino acid identity to SEQ ID NO: 2.
140. The method of Claim 135, wherein said cannabinoid receptor has at least 70% amino acid identity to SEQ ID NO: 2.
141. A method for identifying a compound which binds to a cannabinoid receptor comprising: labeling a compound of Claim 1 with a detectable label; and determining the number of occupied cannabinoid receptors.
142. The method of Claim 141, wherein the detectable label is a radiolabel.
143. The method of Claim 141, wherein the radiolabel is [3H].
PCT/US2006/040662 2005-10-17 2006-10-17 Cb-1 modulating compounds and their use WO2007047737A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP06826157A EP1937677A1 (en) 2005-10-17 2006-10-17 Cb-1 modulating compounds and their use
CA002625423A CA2625423A1 (en) 2005-10-17 2006-10-17 Cb-1 modulating compounds and their use

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US72799705P 2005-10-17 2005-10-17
US60/727,997 2005-10-17
US83100306P 2006-07-14 2006-07-14
US60/831,003 2006-07-14
US83251006P 2006-07-21 2006-07-21
US60/832,510 2006-07-21

Publications (1)

Publication Number Publication Date
WO2007047737A1 true WO2007047737A1 (en) 2007-04-26

Family

ID=37708128

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/040662 WO2007047737A1 (en) 2005-10-17 2006-10-17 Cb-1 modulating compounds and their use

Country Status (4)

Country Link
US (1) US20070105819A1 (en)
EP (1) EP1937677A1 (en)
CA (1) CA2625423A1 (en)
WO (1) WO2007047737A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008017381A1 (en) 2006-08-08 2008-02-14 Sanofi-Aventis Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, processes for preparing them, medicaments comprising these compounds, and their use
WO2008118141A2 (en) * 2006-10-17 2008-10-02 Acadia Pharmaceuticals Inc. Use of cannabinoid modulating compounds in combination with other therapeutic compounds for adjunctive therapy
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010003624A2 (en) 2008-07-09 2010-01-14 Sanofi-Aventis Heterocyclic compounds, processes for their preparation, medicaments comprising these compounds, and the use thereof
WO2010062565A1 (en) * 2008-10-27 2010-06-03 Acadia Pharmaceuticals Inc. Muscarinic agonists
WO2010068601A1 (en) 2008-12-08 2010-06-17 Sanofi-Aventis A crystalline heteroaromatic fluoroglycoside hydrate, processes for making, methods of use and pharmaceutical compositions thereof
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2014112898A1 (en) * 2013-01-16 2014-07-24 Общество С Ограниченной Ответственностью "Биоинтегратор" (Ооо "Биоинтегратор") Conjugates and small molecules which interact with the cd16a receptor
CN103958522A (en) * 2011-11-23 2014-07-30 锡克拜控股有限公司 Polycyclic aromatic compounds containing an S atom or S(=O)2 group and their use as dyes
WO2015017412A1 (en) * 2013-07-29 2015-02-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 11-oxo-10,11-dihydrodibenzo[b,f][1,4]thiazepine s-oxide derivatives and their use as dopamine d2 receptor antagonists
JP2015509077A (en) * 2011-12-09 2015-03-26 リサーチ トライアングル インスティテュート 1-Substituted 4-arylpiperazines as kappa opioid receptor antagonists
US11046651B2 (en) 2019-10-21 2021-06-29 Alairion, Inc. Piperazine substituted azapine derivatives and uses thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1937614A1 (en) * 2005-10-17 2008-07-02 Arcadia Pharmaceuticals Inc. Iron catalyzed cross-coupling reactions of imidoyl derivatives
CN110437153A (en) * 2014-03-13 2019-11-12 美国印第安纳大学研究和技术公司 Hepatitis B virus allosteric modulators
TWI730985B (en) * 2015-09-15 2021-06-21 美商艾森伯利生物科學公司 Hepatitis b core protein modulators
CA3037151A1 (en) 2016-09-15 2018-03-22 Assembly Biosciences, Inc. Hepatitis b core protein modulators
MX2019010404A (en) 2017-03-02 2019-11-21 Assembly Biosciences Inc Cyclic sulfamide compounds and methods of using same.
JP7179751B2 (en) 2017-03-13 2022-11-29 アッセンブリー バイオサイエンシズ,インコーポレイテッド Methods of Making Hepatitis B Core Protein Modulators

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH422805A (en) * 1961-07-20 1966-10-31 Wander Ag Dr A Process for the preparation of amidines
FR1508536A (en) * 1966-01-17 1968-01-05 American Cyanamid Co New process for the preparation of 11-tert-aminodibenz [b, f] [1, 4] oxazepines and thiazepines
EP0040860A1 (en) * 1980-05-28 1981-12-02 Chugai Seiyaku Kabushiki Kaisha Dibenzoxazepine derivative, process for preparing the same and pharmaceutical composition comprising the same
WO2005063254A2 (en) * 2003-12-22 2005-07-14 Acadia Pharmaceuticals Inc. Amino substituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5707798A (en) * 1993-07-13 1998-01-13 Novo Nordisk A/S Identification of ligands by selective amplification of cells transfected with receptors
CN1678303A (en) * 2002-08-23 2005-10-05 康涅狄格大学 Keto cannabinoids with therapeutic indications
EP1937614A1 (en) * 2005-10-17 2008-07-02 Arcadia Pharmaceuticals Inc. Iron catalyzed cross-coupling reactions of imidoyl derivatives
US20080090805A1 (en) * 2005-10-17 2008-04-17 Acadia Pharmaceuticals Inc. Cb-1 modulating compounds and their use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH422805A (en) * 1961-07-20 1966-10-31 Wander Ag Dr A Process for the preparation of amidines
FR1508536A (en) * 1966-01-17 1968-01-05 American Cyanamid Co New process for the preparation of 11-tert-aminodibenz [b, f] [1, 4] oxazepines and thiazepines
EP0040860A1 (en) * 1980-05-28 1981-12-02 Chugai Seiyaku Kabushiki Kaisha Dibenzoxazepine derivative, process for preparing the same and pharmaceutical composition comprising the same
WO2005063254A2 (en) * 2003-12-22 2005-07-14 Acadia Pharmaceuticals Inc. Amino substituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
"ChemDiv, Inc. Product Library", 25 April 2003, CHEMDIV, INC., SAN DIEGO, CA, 92121, USA *
ALLEN, RICHARD C. ET AL: "8-Carboxy-6-sulfamyldibenz[b,f][1,4]oxazepines and -thiazepines as potential high-ceiling diuretics", JOURNAL OF MEDICINAL CHEMISTRY , 21(8), 838-40 CODEN: JMCMAR; ISSN: 0022-2623, 1978, XP002420592 *
DATABASE CHEMCATS CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002420808 *
DATABASE CHEMCATS CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002420809 *
FRYER, R. IAN ET AL: "Quinazolines and 1,4-benzodiazepines. LVII. 1H-1,4-Benzodiazepines", JOURNAL OF HETEROCYCLIC CHEMISTRY , 10(4), 473-8 CODEN: JHTCAD; ISSN: 0022-152X, 1973, XP008075238 *
OTTESEN, LARS K. ET AL: "Iron-Catalyzed Cross-Coupling of Imidoyl Chlorides with Grignard Reagents - SUPPORTING INFORMATION", 30 March 2006 (2006-03-30), pages S1 - S15, XP002420597, Retrieved from the Internet <URL:http://pubs.acs.org/subscribe/journals/orlef7/suppinfo/ol0600234/ol0600234si20060317_035200.pdf> [retrieved on 20070216] *
OTTESEN, LARS K. ET AL: "Iron-Catalyzed Cross-Coupling of Imidoyl Chlorides with Grignard Reagents", ORGANIC LETTERS , 8(9), 1771-1773 CODEN: ORLEF7; ISSN: 1523-7060, 2006, XP002420596 *
SUNDRAM ET AL: "Clozapine decreases [3H] CP 55940 binding to the cannabinoid1 receptor in the rat accumbens", NANYN-SCHMIEDEBERG'S ARCH. PHARMACOL., vol. 371, 2 July 2005 (2005-07-02), pages 428 - 433, XP002420595 *
T. C. MCKENZIE: "The reductive methylation of dibenzo[b,f][1,4]oxazepines", SYNTHESIS, 1983, pages 288 - 289, XP002420594 *
UMEMIYA ET AL: "Regulation of Retinoidal Actions by Diazepinylbenzoic Acids. Retinoid Synergists which activate RXR-RAR Heterodimers", J. MED. CHEM., vol. 40, 1997, pages 4222 - 4234, XP002420593 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008017381A1 (en) 2006-08-08 2008-02-14 Sanofi-Aventis Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, processes for preparing them, medicaments comprising these compounds, and their use
WO2008118141A2 (en) * 2006-10-17 2008-10-02 Acadia Pharmaceuticals Inc. Use of cannabinoid modulating compounds in combination with other therapeutic compounds for adjunctive therapy
WO2008118141A3 (en) * 2006-10-17 2008-12-24 Acadia Pharm Inc Use of cannabinoid modulating compounds in combination with other therapeutic compounds for adjunctive therapy
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010003624A2 (en) 2008-07-09 2010-01-14 Sanofi-Aventis Heterocyclic compounds, processes for their preparation, medicaments comprising these compounds, and the use thereof
WO2010062565A1 (en) * 2008-10-27 2010-06-03 Acadia Pharmaceuticals Inc. Muscarinic agonists
WO2010068601A1 (en) 2008-12-08 2010-06-17 Sanofi-Aventis A crystalline heteroaromatic fluoroglycoside hydrate, processes for making, methods of use and pharmaceutical compositions thereof
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
CN103958522A (en) * 2011-11-23 2014-07-30 锡克拜控股有限公司 Polycyclic aromatic compounds containing an S atom or S(=O)2 group and their use as dyes
JP2015509077A (en) * 2011-12-09 2015-03-26 リサーチ トライアングル インスティテュート 1-Substituted 4-arylpiperazines as kappa opioid receptor antagonists
US9512105B2 (en) 2011-12-09 2016-12-06 Research Triangle Institute 1-substituted 4-arylpiperazine as kappa opioid receptor antagonists
JP2016505629A (en) * 2013-01-16 2016-02-25 アラ・ケム・エルエルシー Complexes and small molecules that interact with the CD16a receptor
CN104870428A (en) * 2013-01-16 2015-08-26 生物集成有限责任公司 Conjugates and small molecules which interact with the CD16A receptor
WO2014112898A1 (en) * 2013-01-16 2014-07-24 Общество С Ограниченной Ответственностью "Биоинтегратор" (Ооо "Биоинтегратор") Conjugates and small molecules which interact with the cd16a receptor
AU2014207918B2 (en) * 2013-01-16 2017-06-08 "Biointegrator" Limited Liability Company (Ooo "Biointegrator") Conjugates and small molecules which interact with the CD16a receptor
EA028617B1 (en) * 2013-01-16 2017-12-29 Общество С Ограниченной Ответственностью "Биоинтегратор" (Ооо "Биоинтегратор") CONJUGATES AND SMALL MOLECULES WHICH INTERACT WITH THE CD16a RECEPTOR
WO2015017412A1 (en) * 2013-07-29 2015-02-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 11-oxo-10,11-dihydrodibenzo[b,f][1,4]thiazepine s-oxide derivatives and their use as dopamine d2 receptor antagonists
US9550742B2 (en) 2013-07-29 2017-01-24 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 11-OXO-10,11-dihydrodibenzo[B,F][1,4]thiazepine S-oxide derivatives and their use as dopamine D2 receptor antagonists
US11046651B2 (en) 2019-10-21 2021-06-29 Alairion, Inc. Piperazine substituted azapine derivatives and uses thereof

Also Published As

Publication number Publication date
US20070105819A1 (en) 2007-05-10
CA2625423A1 (en) 2007-04-26
EP1937677A1 (en) 2008-07-02

Similar Documents

Publication Publication Date Title
WO2007047737A1 (en) Cb-1 modulating compounds and their use
WO2008118141A2 (en) Use of cannabinoid modulating compounds in combination with other therapeutic compounds for adjunctive therapy
CN102264707B (en) Bicyclic heterocyclic compound
AU2015370588B2 (en) Fused bicyclic compounds for the treatment of disease
WO2008048648A2 (en) Cb1-modulating compounds and their use
KR20050085691A (en) 4,5-diarylthiazole derivatives as cb-1 ligands
TWI557110B (en) Arylsulfonyl pyrazoline carboxamidine derivatives as 5-ht6 antagonists
AU2008345681A1 (en) Imidazo [1,2-a] pyridine compounds
EP0091241A2 (en) Condensed pyrrolinone derivatives, and their production
US5162340A (en) Substituted 1-(2h)-isoquinolinones bearing acidic functional groups as angiotensin ii antagonists
WO2006018309A1 (en) 5-ht7 receptor antagonists
EP1057814B1 (en) Tetrahydrobenzindole derivatives
US5173486A (en) Dibenz[b,f][1,4]oxazepin-11(10H)-ones for multidrug resistance reversing agents
WO2009075691A1 (en) Cb-1 modulating compounds and their use
EP1725232B1 (en) (indol-3-yl)-heterocycle derivatives as agonists of the cannabinoid cb1 receptor
KR20090086641A (en) Tricyclic compound and medical use thereof
KR20090023701A (en) Tricyclic compound and pharmaceutical use thereof
WO2006018308A1 (en) 5-ht7 receptor antagonists
JP2009057281A (en) P2x4 receptor antagonist
EP1100803B1 (en) Pyrrolo [2,1-b] [1,3]benzothiazepines with atypical antipsychotic activity
PL183712B1 (en) Prokinetic oxadiazoles
KR100521959B1 (en) New 2,3-benzodiazepine derivatives
JPH0616638A (en) Pyridine compound and its medicinal use
WO2008061029A1 (en) Aryl sulfonamide derivatives and methods of their use
JP2002201172A (en) Benzamide derivative and its application

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006826157

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2625423

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008536754

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: JP