WO2012090177A2 - Modulateurs des récepteurs cannabinoïdes - Google Patents

Modulateurs des récepteurs cannabinoïdes Download PDF

Info

Publication number
WO2012090177A2
WO2012090177A2 PCT/IB2011/056007 IB2011056007W WO2012090177A2 WO 2012090177 A2 WO2012090177 A2 WO 2012090177A2 IB 2011056007 W IB2011056007 W IB 2011056007W WO 2012090177 A2 WO2012090177 A2 WO 2012090177A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino
methanone
methylisoquinolin
piperidin
morpholino
Prior art date
Application number
PCT/IB2011/056007
Other languages
English (en)
Other versions
WO2012090177A3 (fr
Inventor
Sanjeev Anant Kulkarni
Sachin MADAN
Nirmal Kumar JANA
Prashant Vitthalrao TALE
Narasimha Murthy CHEEMALA
Sachin Jaysing Mahangare
Prashant Popatrao VIDHATE
Chaitanya Prabhakar KULKARNI
Sapana Suresh PATEL
Amolsing Dattu PATIL
Rohan Mahadev SHINDE
Venkata P. Palle
Rajender Kumar Kamboj
Original Assignee
Lupin Limited
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 Lupin Limited filed Critical Lupin Limited
Priority to IN226MUN2014 priority Critical patent/IN2014MN00226A/en
Priority to PCT/IB2012/053407 priority patent/WO2013005168A2/fr
Priority to US13/542,234 priority patent/US9006442B2/en
Publication of WO2012090177A2 publication Critical patent/WO2012090177A2/fr
Publication of WO2012090177A3 publication Critical patent/WO2012090177A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/48Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring being part of a condensed ring system of the same carbon skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/52Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring condensed with a ring other than six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/40Acylated substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/44Radicals substituted by doubly-bound oxygen, sulfur, or nitrogen atoms, or by two such atoms singly-bound to the same carbon atom
    • C07D213/53Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • C07D217/24Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • C07D217/26Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/301,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings
    • 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
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/04Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems

Definitions

  • the present invention relates to compounds, pharmaceutically acceptable salts thereof and pharmaceutical compositions for the treatment, prevention, management, and/or lessening the severity of diseases, disorders, syndromes or conditions associated with the modulation of cannabinoid (CB) receptors.
  • the invention also relates to methods of treating, preventing, managing and/or lessening the severity of the diseases disorders, syndromes or conditions associated with the modulation of cannabinoid (CB) receptors.
  • the invention also relates to processes for the preparation of the compounds of the invention.
  • CX5 G-protein coupled receptor expressed in human promyelocytic leukemic cell line (HL60) was discovered in 1993. This was later identified as type I I Cannabinoid receptor (Munro et al. Nature (1993), 365, 61 -65). CB 2 receptors are found to be expressed predominantly in immune cells. When activated they modulate immune cell migration and cytokine release outside and within the brain (Pertwee et al. Handb. Exp.
  • CB 2 activation affects a host of immune responses from inflammation to neuroprotection (Cabral et al. Handb. Exp. Pharmacol. (2005), 168, 385- 423).
  • the activation of CB 2 receptors is reported to have analgesic effect in many animal models of pain from acute pain to neuropathic pain (Anand et al. Pain. (2009), 138, 667-
  • CB 2 is an attractive therapeutic target for pain management and immune system modulation without overt psycho activity and substance abuse possibility, its presumed absence in the CNS was reviewed by many researchers. The findings of these recent investigations support the presence of CB 2 receptors in CNS.
  • the CB 2 imRNA and protein have been found in microglia (Beltramo et al. Eur. J. Neurosci. (2006), 23, 1530-1 538; Carlisle et al. Int. Immuno Pharmacol. (2002), 2, 69-82; Klegeris et al. Br. J. Pharmacol. (2003), 139, 775-786; Maresz et al., Nat. Med. (2007), 13, 492-497; Walter et al. J. Neurosci.
  • CB 2 The expression levels of CB 2 are proportional to the activation extent of microglia (Cabral et al., Br. J. Pharmacol. (2008), 153, 240-251 ; Pietr et al. FEBS Lett. (2009), 583, 2071 -2076; Stella et al. Gila. (2004), 48, 267-277). Microglial migration and their infiltration into brain areas with active neuroinflammation and degeneration is modulated by CB 2 . In healthy brain microglia does not express CB 2 but they do in Alzheimer's brain tissue in the neuritic-plaque associated microglia (Benito et al. J Neurosci.
  • CB 2 imRNA is found to increase in association with activated microglia in the spinal cord (Zhang et al. Eur. J. Neurosci. (2003), 17, 2750-2754). Similarly in amyotrophic lateral sclerosis and multiple sclerosis, CB 2 microglial expression increases in spinal cord (Yiangou et al. BMC. Neurol. (2006), 6, 12).
  • CB 2 receptor distribution is also known in macrophages, CD4 T cells, CD8 T cells, B cells, natural killer cells, monocytes, and polymorphonuclear neutrophils (Dittel B N. et al. BJP. (2008), 153, (2), 271 -276; Maresz K. et al. Nature Medicine. (2007), 13, 492-497).
  • CB 2 receptor presence is also known in other brain areas like thalamus, straitum, hippocampus.
  • Peripheral neurons, nociceptive neurons and sensory neurons also are known to express CB 2 receptors.
  • CB 2 receptor is expressed in the bone cells and is reported to have anabolic effect on bones (Ofek et al. PNAS. (2006), 103, 696-701 ). Presence of CB 2 is also reported in the enteric nervous system, (Duncan M Mouihate et al, Am. J. Physiol. Gastrointest. Liver Physiol. 295, G78-G87). Similar anabolic effect is also reported through CB2 receptor mediation in neurogenesis (Molina-Holgado et al. Eur. J. Neurosci. (2007), 25, 629-634 & Palazuelos J. et al FASEB J. (2006), 20, 2405-2407).
  • CB1 in pain management is also well known. This receptor is known to exist majorly in the central nervous system. To a minor extent, CB1 is also reported to exist in the periphery (Pharmacology, Biochemistry and Behavior 90 (2008) 501 -51 1 ). Therefore, along with compounds that are CB2 selective modulators, dual modulators working through CB1 and CB2 are also targeted.
  • WO 2002/042248, WO 2010/096371 , WO 201 0/133973, WO 2006/129178, WO 201 0/ 084767, WO 2009/053799, WO 2008/1 1 5672, WO 2008/027812, WO 2007/102059, WO 2007/091950 and WO 2002/42248 applications disclose compounds related to cannabinoid receptors for the treatment of various diseases mediated by cannabinoid receptors. Also, Bioorganic Medicinal Chemistry, (201 1 ), IjL. 939-950, discloses the compounds related to cannabinoid receptors.
  • the invention provides compounds having the structure of Formula (I) :
  • W is -N- or -CH-
  • X is selected from -NR a -, -0- and -S(0) n -;
  • Y is selected from -C(O)-, -S(0) 2 -, and -CR 5 R 6 -;
  • R which may be same or different at each occurrence, is independently selected from halogen, cyano, nitro, hydroxy, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl , alkoxy, cycloalkoxy, -C(0)OH, -NR a R b and -C(0)NR a R b ;
  • Ri is selected from hydrogen, alkyl, cycloalkyl, halogen, hydroxy, cyano, amino, nitro, alkoxy, haloalkyl, haloalkoxy and cycloalkoxy;
  • R 2 is selected from hydrogen , alkyl, cycloalkyl , cycloalkyl alkyl , aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ;
  • R 3 is selected from alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ; or
  • R 2 and R 3 together with the nitrogen atom to which they are attached, may form a substituted or an unsubstituted 3 to 1 4 membered heterocyclic ring ;
  • R 2 and R 3 together with the nitrogen atom to which they are attached, may form a substituted or an unsubstituted 5 to 1 4 membered heteroaryl ring ;
  • R 4 is selected from cycloalkyl , aryl, heteroaryl and heterocyclyl ;
  • R 5 and R 6 which may be same or different, are independently selected from hydrogen, alkyl and haloalkyl ;
  • R a and R b which may be same or different at each occurrence, are independently selected from hydrogen , alkyl, haloalkyi, acyl , cycloalkyi and aryl ; or R a and R b , together with the nitrogen atom to which they are attached, may form a substituted or unsubstituted 3 to 1 4 membered heteroaryl or heterocyclic ring ;
  • 'm' is an integer ranging from 0 to 3, both inclusive;
  • 'n' is an integer ranging from 0 to 2, both inclusive;
  • R is halogen, alkyl or cycloalkyi ;
  • R 7 which may be same or different at each occurrence, is independently selected from halogen, cyano, nitro, hydroxy, alkyl, haloalkyi, hydroxyalkyi, cycloalkyi , alkoxy, haloalkoxy, cycloalkoxy, -C(0)OH, -NR a R b and -C(0)NR a R b ;
  • R 2 is selected from hydrogen , alkyl, cycloalkyl , cycloalkylalkyl , aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ;
  • R 3 is selected from alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ;
  • R a and R b are as defined herein above;
  • 'p' is an integer ranging from 0 to 4, both inclusive;
  • ring A is a substituted or unsubstituted 3 to 14 membered heterocyclic ring wherein substituents on ring A may be on same or different ring atom ;
  • Y is-C(0)- or -CH 2 -;
  • Ri is halogen, alkyl or cycloalkyl ;
  • R 7 which may be same or different at each occurrence, is independently selected from halogen, cyano, nitro, hydroxy, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl , alkoxy, haloalkoxy, cycloalkoxy, -C(0)OH, -NR a R b and -C(0)NR a R b ;
  • R a and R b are as defined herein above;
  • 'p' is an integer ranging from 0 to 4, both inclusive;
  • R 7 which may be same or different at each occurrence, is independently selected from halogen, cyano, nitro, hydroxy, alkyl , haloalkyl , hydroxyalkyl, cycloalkyl, alkoxy, haloalkoxy, cycloalkoxy, -C(0)OH, -NR a R b and -C(0)N R a R b ;
  • R 2 is selected from hydrogen , alkyl, cycloalkyl , cycloalkyl alkyl , aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ;
  • R 3 is selected from alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ; or
  • R 2 and R 3 together with the nitrogen atom to which they are attached, may form a substituted or unsubstituted 3 to 1 4 membered heterocyclic ring ;
  • R a and R b are as defined herein above;
  • 'p' is an integer ranging from 0 to 4, both inclusive;
  • Ri is halogen, alkyl or cycloalkyl ;
  • R 7 which may be same or different at each occurrence, is independently selected from halogen, cyano, nitro, hydroxy, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, alkoxy, haloalkoxy, cycloalkoxy, -C(0)OH, -NR a R b and -C(0)NR a R b ;
  • R 2 is selected from hydrogen , alkyl, cycloalkyl , cycloalkylalkyl , aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl ;
  • R 3 is selected from alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl; or
  • R 2 and R 3 together with the nitrogen atom to which they are attached, may form a substituted or unsubstituted 3 to 14 membered heterocyclic ring ;
  • R a and R b are as defined herein above;
  • 'p' is an integer ranging from 0 to 4, both inclusive;
  • Ri is halogen, alkyl or cycloalkyl
  • R 7 which may be same or different at each occurrence, is independently selected from halogen, cyano, nitro, hydroxy, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, alkoxy, haloalkoxy, cycloalkoxy, -C(0)OH, -NR a R b and -C(0)NR a R b ;
  • R 2 is selected from hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl;
  • R 3 is selected from alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl; or
  • R 2 and R 3 together with the nitrogen atom to which they are attached, may form a substituted or unsubstituted 3 to 14 membered heterocyclic ring ;
  • R a and R b are as defined herein above;
  • 'p' is an integer ranging from 0 to 4, both inclusive;
  • formulae (I), (II), (I II) (IV), (V) or (VI) structurally encompasses all N-oxides, tautomers, stereoisomers including isotopes wherever applicable and pharmaceutically acceptable salts that may be contemplated from the chemical structure of the genera described herein.
  • heterocyclic ring is monocyclic (for example azetidine, pyrrolidine, piperidine, piperazine, morpholine, dioxidothiomorpholine or tetrahydropyran) fused or bridged bicyclic (for example 6-oxa-3-azabicyclo(3.1 .1 )heptane, 2-oxa-5- azabicyclo[2.2.1 ]heptane, 3-azabicyclo[3.1 .0]hexane or 3-oxa-9- azabicyclo[3.3.1 ]nonane or spirocyclic for example 2-oxa-6-azaspiro[3.3]heptane, 2-oxa- 6-azaspiro[3.5]nonane, 2-oxa-7-azaspiro[3.5
  • ring A is substituted or unsubstituted 3 to 12 membered heterocyclic ring wherein the heterocyclic ring is monocyclic (for example azetidine, pyrrolidine, piperidine, piperazine or morpholine, tetrahydropyran) fused or bridged bicyclic (for example 6-oxa- 3-azabicyclo[3.1 .1 ]heptane, 2-oxa-5-azabicyclo[2.2.1 ]heptane, 3- azabicyclo[3.1 .0]hexane or 3-oxa-9-azabicyclo[3.3.1 ]nonane or spirocyclic for example 2-oxa-6-azaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.5]nonane, 2-oxa-7-azaspiro[3.5] nonane.
  • monocyclic for example azetidine, pyrrolidine, piperidine, piperazine or morpholine,
  • compounds of formulae (I I), (V) and/or (VI) in which is substituted or unsubstituted alkyl (for example methyl or ethyl) or cycloalkyi for example cyclopropyl ;
  • R 7 is halogen, hydroxy, alkyl or alkoxy;
  • 'p' is 0, 1 , 2 or 3; and one of R 2 and R 3 is hydrogen and the other is selected from alkyl, arylalkyl, heteroarylalkyl, heterocyclyl and heterocyclylalkyl .
  • Ri is substituted or unsubstituted alkyl (for example methyl or ethyl) or cycloalkyl for example cyclopropyl ;
  • R 7 is halogen, hydroxy, alkyl or alkoxy;
  • 'p' is 0, 1 , 2 or 3; and
  • R 2 and R 3 together with nitrogen atom to which they are attached, may form a substituted or unsubstituted 3 to 12 membered heterocyclic ring wherein the heterocyclic ring may be mono or bicyclic, fused, bridged or spirocyclic ring.
  • Y is -C(O)- or -CH 2 -; is substituted or unsubstituted alkyl (for example methyl or ethyl) or cycloalkyl for example cyclopropyl ;
  • R 7 is halogen, hydroxy, alkyl or alkoxy;
  • 'p' is 0, 1 , 2 or 3;
  • ring A is monocyclic (for example azetidine, pyrrolidine, piperidine, piperazine or morpholine, tetrahydropyran) fused or bridged bicyclic (for example 6-oxa-3- azabicyclo[3.1 .1 ]heptane, 2-oxa-5-azabicyclo [2.2.1 ]heptane, 3-azabicyclo[3.1 .0]hexane or 3-oxa-9-azabicyclo[3.3.1 ]nonane or spirocyclic for example 2-oxa-6-;
  • R 7 is halogen, hydroxy, alkyl or alkoxy; 'p' is 0, 1 , 2 or 3; and one of R 2 and R 3 is hydrogen and the other is selected from alkyl, arylalkyl, heteroarylalkyl, heterocyclyl and h etero cycl y I a I ky I ; or R 2 and R 3 together with nitrogen atom to which they are attached, may form a substituted or unsubstituted 3 to 12 membered heterocyclic ring wherein the heterocyclic ring may be monocyclic (for example azetidine, pyrrolidine, piperidine, piperazine or morpholine, dioxidothiomorpholine, tetrahydropyran) fused or bridged bicyclic (for example 6-oxa-3-azabicyclo[3.1 .1 ]heptane, 2-oxa-5- azabicyclo[2.2.1 ]heptan
  • a compound of formula (I) useful in treating , preventing , managing and/or lessening the severity of diseases, disorders, syndromes or conditions associated with cannabinoid (CB) modulators.
  • the invention provides a pharmaceutical composition that includes at least one compound of formula (I) and at least one pharmaceutically acceptable excipient.
  • the invention provides a pharmaceutical composition of compound of formula (I) useful in treating, preventing, managing and/or lessening the severity of the diseases, disorders, syndromes or conditions associated with cannabinoid (CB) modulators in a subject, in need thereof by administering to the subject, one or more compounds described herein in a therapeutically effective amount to cause modulation of such receptor.
  • CBD cannabinoid
  • halogen or halo means fluorine, chlorine, bromine, or iodine.
  • acyl refers to a group or radical derived from carboxylic acid for example alkylcarbonyl (for example CH 3 C(0)-) or arylcarbonyl. Unless set forth or recited to the contrary, all acyl groups described or claimed herein may be substituted or unsubstituted.
  • alkyl refers to an alkane derived hydrocarbon radical that includes solely carbon and hydrogen atoms in the backbone, contains no unsaturation, has from one to six carbon atoms, and is attached to the remainder of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1 -methylethyl (isopropyl), n-butyl, n-pentyl, 1 ,1 - dimethylethyl (t-butyl) and the like. Unless set forth or recited to the contrary, all alkyl groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • alkenyl refers to a hydrocarbon radical containing from 2 to 10 carbon atoms and including at least one carbon-carbon double bond.
  • alkenyl groups include ethenyl, 1 -propenyl, 2-propenyl (allyl), /so-propenyl, 2-methyl-l- propenyl, 1 -butenyl, 2-butenyl and the like. Unless set forth or recited to the contrary, all alkenyl groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • alkynyl refers to a hydrocarbon radical containing 2 to 10 carbon atoms and including at least one carbon- carbon triple bond.
  • Non- limiting examples of alkynyl groups include ethynyl, propynyl, butynyl and the like. Unless set forth or recited to the contrary, all alkynyl groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • alkoxy refers to an alkyl group attached via an oxygen linkage. Non-limiting examples of such groups are methoxy, ethoxy and propoxy and the like. Unless set forth or recited to the contrary, all alkoxy groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • cycloalkyi refers to a non-aromatic mono or multicyclic ring system having 3 to 12 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • multicyclic cycloalkyi groups include, but are not limited to, perhydronapththyl, adamantyl and norbornyl groups, bridged cyclic groups or spirobicyclic groups, e.g., spiro(4,4)non-2-yl and the like. Unless set forth or recited to the contrary, all cycloalkyi groups described or claimed herein may be substituted or unsubstituted.
  • cycloalkoxy refers to an cycloalkyl, defined herein, group attached via an oxygen linkage.
  • Non-limiting examples of such groups are cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy and the like. Unless set forth or recited to the contrary, all alkoxy groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • cycloalkenyl refers to a non-aromatic mono or multicyclic ring system having 3 to 12 carbon atoms and including at least one carbon-carbon double bond, such as cyclopentenyl, cyclohexenyl, cycloheptenyl and the like. Unless set forth or recited to the contrary, all cycloalkenyl groups described or claimed herein may be substituted or unsubstituted.
  • cycloalkylalkyi refers to a cycloalkyl group as defined above, directly bonded to an alkyl group as defined above, e.g., cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, cyclohexylethyl, etc. Unless set forth or recited to the contrary, all cycloalkylalkyi groups described or claimed herein may be substituted or unsubstituted.
  • haloalkyi refers to an alkyl group as defined above that is substituted by one or more halogen atoms as defined above.
  • the haloalkyi may be monohaloalkyl, dihaloalkyl or polyhaloalkyl including perhaloalkyl.
  • a monohaloalkyl can have one iodine, bromine, chlorine or fluorine atom.
  • Dihaloalkyl and polyhaloalkyl groups can be substituted with two or more of the same halogen atoms or a combination of different halogen atoms.
  • a polyhaloalkyl is substituted with up to 12 halogen atoms.
  • Non-limiting examples of a haloalkyi include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl, dichloropropyl and the like.
  • a perhaloalkyl refers to an alkyl having all hydrogen atoms replaced with halogen atoms.
  • haloalkoxy refers to a haloalkyi, defined herein, group attached via an oxygen linkage.
  • Non-limiting examples of such groups are monohaloalkoxy, dihaloalkoxy or polyhaloalkoxy including perhaloalkoxy.
  • all haloalkoxy groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • hydroxyalkyi refers to an alkyl group, as defined above that is substituted by one or more hydroxy groups.
  • the hydroxyalkyi is monohydroxyalkyl or dihydroxyalkyl.
  • Non-limiting examples of a hydroxyalkyl include 2- hydroxyethyl, 3- hydroxypropyl, 2-hydroxypropyl, and the like.
  • aryl refers to an aromatic radical having 6- to 14- carbon atoms, including monocyclic, bicyclic and tricyclic aromatic systems, such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, and biphenyl and the like. Unless set forth or recited to the contrary, all aryl groups described or claimed herein may be substituted or unsubstituted.
  • arylalkyl refers to an aryl group as defined above directly bonded to an alkyl group as defined above, e.g., -CH 2 C 6 H 5 and -C 2 H 4 C 6 H5. Unless set forth or recited to the contrary, all arylalkyl groups described or claimed herein may be substituted or unsubstituted.
  • heterocyclic ring or “heterocyclyl ring” or “heterocyclyl”, unless otherwise specified, refers to substituted or unsubstituted non-aromatic 3- to 15- membered ring which consists of carbon atoms and with one or more heteroatom(s) independently selected from N, O or S.
  • the heterocyclic ring may be a mono-, bi- or tricyclic ring system, which may include fused, bridged or spiro ring systems and the nitrogen, carbon, oxygen or sulfur atoms in the heterocyclic ring may be optionally oxidized to various oxidation states.
  • heterocyclic ring may also be fused with aromatic ring.
  • heterocyclic rings include azepinyl, azetidinyl, benzodioxolyl, benzodioxanyl, benzopyranyl, chromanyl, dioxolanyl, dioxaphospholanyl, decahydroisoquinolyl, indanyl, indolinyl, isoindolinyl, isochromanyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, oxazolinyl, oxazolidinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2- oxoazepinyl, octahydroindolyl, octahydroisoindolyl, perhydroazepinyl, piperazinyl, 4- piperidonyl, pyrrolidinyl, piperidiny
  • heterocyclic ring may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure. Unless set forth or recited to the contrary, all heterocyclyl groups described or claimed herein may be substituted or unsubstituted; substituents may be on same or different ring atom.
  • heteroaryl refers to a substituted or unsubstituted 5- to 14- membered aromatic heterocyclic ring with one or more heteroatom(s) independently selected from N, O or S.
  • the heteroaryl may be a mono-, bi- or tricyclic ring system.
  • the heteroaryl ring may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure.
  • Non-limiting examples of a heteroaryl ring include oxazolyl, isoxazolyl, imidazolyl, furyl, indolyl, isoindolyl, pyrrolyl, triazolyl, triazinyl, tetrazolyl, thienyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzofuranyl, benzothiazolyl, benzoxazolyl, benzimidazolyl, benzothienyl, carbazolyl, quinolinyl, isoquinolinyl, quinazolinyl, cinnolinyl, naphthyridinyl, pteridinyl, purinyl, quinoxalinyl, quinolyl, isoquinolyl, thiadiazolyl, indolizinyl, acridinyl
  • heterocyclylalkyi refers to a heterocyclic ring radical directly bonded to an alkyl group.
  • the heterocyclylalkyi radical may be attached to the main structure at any carbon atom in the alkyl group that results in the creation of a stable structure. Unless set forth or recited to the contrary, all heterocyclylalkyi groups described or claimed herein may be substituted or unsubstituted.
  • heteroarylalkyl refers to a heteroaryl ring radical directly bonded to an alkyl group.
  • the heteroarylalkyl radical may be attached to the main structure at any carbon atom in the alkyl group that results in the creation of a stable structure. Unless set forth or recited to the contrary, all heteroarylalkyl groups described or claimed herein may be substituted or unsubstituted.
  • substituted refers to a group or moiety having one or more substituents attached to the structural skeleton of the group or moiety.
  • phrases “may optionally be substituted” refers to a moiety or group that may or may not be substituted.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted and unsubstituted aryl radicals.
  • a “stereoisomer” refers to a compound having the same atoms bonded through the same bonds but having different three-dimensional orientations, which are not interchangeable.
  • the invention contemplates various stereoisomers and mixtures thereof and includes enantiomers and diastereomers.
  • the invention also includes geometric isomers "E” or “Z” or cis or trans configuration in a compound having either a double bond or having substituted cycloalkyl ring system.
  • a “tautomer” refers to a compound that undergoes rapid proton shifts from one atom of the compound to another atom of the compound. Some of the compounds described herein may exist as tautomers with different points of attachment of hydrogen. The individual tautomers as well as mixture thereof are encompassed with compounds of formula (I).
  • treating or “treatment” of a state, disease, disorder, condition or syndrome includes: (a) preventing or delaying the appearance of clinical symptoms of the state, disease, disorder, condition or syndrome developing in a subject that may be afflicted with or predisposed to the state, disease, disorder, condition or syndrome but does not yet experience or display clinical or subclinical symptoms of the state, disease, disorder, condition or syndrome; (b) inhibiting the state, disease, disorder, condition or syndrome, i.e., arresting or reducing the development of the disease or at least one clinical or subclinical symptom thereof; c) lessening the severity of a disease, disorder, syndrome or condition or at least one of its clinical or subclinical symptoms thereof; and/or (d) relieving the disease, i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms.
  • modulate refers to an increase or decrease in the amount, quality, or effect of a particular activity, function or molecule. By way of illustration and not limitation, it includes agonists, partial agonists, inverse agonists and antagonists of a cannabinoid (CB) receptor of the present invention.
  • CB cannabinoid
  • subject includes mammals, preferably humans and other animals, such as domestic animals; e.g., household pets including cats and dogs.
  • a “therapeutically effective amount” refers to the amount of a compound that, when administered to a subject in need thereof, is sufficient to cause a desired effect.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity, age, weight, physical condition and responsiveness of the subject to be treated.
  • the compounds of the invention may form salts.
  • administration of the compound as a pharmaceutically acceptable salt may be appropriate.
  • pharmaceutically acceptable salts are organic acid addition salts formed by addition of acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, a-glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, and salicylate.
  • Suitable inorganic salts may also be formed, including, sulfate, nitrate, bicarbonate, carbonate salts, hydrobromate and phosphoric acid.
  • salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • the invention extends to stereoisomeric forms and to mixtures thereof.
  • the different stereoisomeric forms of the invention may be separated from one another by the method known in the art, or a given isomer may be obtained by stereospecific or asymmetric synthesis.
  • Tautomeric forms and mixtures of compounds described herein are also contemplated. Screening of compounds of invention for cannabinoid receptor modulation activity can be achieved by using various in vitro and in vivo protocols mentioned herein below or methods known in the art.
  • compositions The invention relates to pharmaceutical composition containing the compounds of the Formula (I) disclosed herein.
  • pharmaceutical compositions containing a therapeutically effective amount of at least one compound of formula (I) described herein and at least one pharmaceutically acceptable excipient (such as a carrier or diluent).
  • the contemplated pharmaceutical compositions include the compound(s) described herein in an amount sufficient to modulate cannabinoid receptor mediated diseases described herein when administered to a subject.
  • the subjects contemplated include, for example, a living cell and a mammal, including human mammal.
  • the compound of the invention may be associated with a pharmaceutically acceptable excipient (such as a carrier or a diluent) or be diluted by a carrier, or enclosed within a carrier which can be in the form of a capsule, sachet, paper or other container.
  • a pharmaceutically acceptable excipient includes a pharmaceutical agent that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • suitable carriers include, but are not limited to, water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicylic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone.
  • the pharmaceutical composition may also include one or more pharmaceutically acceptable auxiliary agents, wetting agents, emulsifying agents, suspending agents, preserving agents, salts for influencing osmotic pressure, buffers, sweetening agents, flavoring agents, colorants, or any combination of the foregoing.
  • the pharmaceutical composition of the invention may be formulated so as to provide quick, sustained, or delayed release of the active ingredient after administration to the subject by employing procedures known in the art.
  • the pharmaceutical compositions described herein may be prepared by conventional techniques known in the art.
  • the active compound can be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier, which may be in the form of an ampoule, capsule, sachet, paper, or other container.
  • the carrier serves as a diluent, it may be a solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound.
  • the active compound can be adsorbed on a granular solid container, for example, in a sachet.
  • the pharmaceutical compositions may be in conventional forms, for example, capsules, tablets, aerosols, solutions, suspensions or products for topical application.
  • the route of administration may be any route which effectively transports the active compound of the invention to the appropriate or desired site of action.
  • Suitable routes of administration include, but are not limited to, oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal, parenteral, rectal, depot, subcutaneous, intravenous, intraurethral, intramuscular, intranasal, ophthalmic (such as with an ophthalmic solution) or topical (such as with a topical ointment).
  • Solid oral formulations include, but are not limited to, tablets, capsules (soft or hard gelatin), dragees (containing the active ingredient in powder or pellet form), troches and lozenges. Tablets, dragees, or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application.
  • Liquid formulations include, but are not limited to, syrups, emulsions, soft gelatin and sterile injectable liquids, such as aqueous or non-aqueous liquid suspensions or solutions.
  • injectable solutions or suspensions formulation include, but are not limited to, syrups, emulsions, suspensions, solutions, emulsions, soft gelatin and sterile injectable liquids, such as aqueous or non-aqueous liquid suspensions or solutions.
  • injectable solutions or suspensions preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil.
  • the pharmaceutical preparation is preferably in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as pocketed tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • the total daily dose of the compounds of the invention depends, of course, on the mode of administration.
  • oral administration may require a higher total daily dose, than an intravenous (direct into blood).
  • the quantity of active component in a unit dose preparation may be varied or adjusted from 0.1 mg to 1 0000 mg according to the potency of the active component or mode of administration.
  • Suitable doses of the compounds for use in treating the diseases and disorders described herein can be determined by those skilled in the relevant art.
  • Therapeutic doses are generally identified through a dose ranging study in subject based on preliminary evidence derived from the animal studies. Doses must be sufficient to result in a desired therapeutic benefit without causing unwanted side effects for the patient.
  • the daily dosage of the CB modulator can range from about 0.1 to about 30.0 mg/kg.
  • Mode of administration, dosage forms, suitable pharmaceutical excipients, diluents or carriers can also be well used and adjusted by those skilled in the art. All changes and modifications are envisioned within the scope of the invention.
  • the invention provides compounds and pharmaceutical compositions thereof that are useful in treating, preventing, managing and/or lessening the severity of diseases, disorders, syndromes or conditions modulated by cannabinoid receptor.
  • the invention further provides method of treating diseases, disorders or conditions modulated by cannabinoid receptor in a subject in need thereof by administering to the subject a therapeutically effective amount of a compound or a pharmaceutical composition of the invention.
  • the methods provided are also useful for diagnosis of conditions that can be treated by acting on cannabinoid receptor for determining if a patient will be responsible to therapeutic agents.
  • the invention provides a method for the treatment of diseases, disorders or conditions through modulating cannabinoid receptor.
  • a subject in need of such treatment is administered a therapeutically effective amount of a compound of formula (I) described herein.
  • the compound of formula (I), being modulators of cannabinoid receptor, is potentially useful in treating, preventing, managing and/or lessening the severity of diseases, disorders, syndromes or conditions include but are not limited to pain, inflammation, analgesic conditions, healing of wounds and burns, movement disorders, immune disorders (such as autoimmune disorders), respiratory disorders, lung diseases associated with inflammation, pruritis associated with inflammation, allergic diseases associated with inflammation, organ contraction, preanesthetic medication, pre operative medication, muscle spasm, locomotor activity disorders, bone disorders, multiple sclerosis, glaucoma and related intra ocular pressure, cell growth disorders, gastrointestinal disorders, diseases of central nervous system (CNS) erythromyalgia, neurological disorders, neurodegenerative disorders, neuromuscular conditions, neuroinflammatory pathologies and the like.
  • CNS central nervous system
  • Pain includes, but is not limited to, acute pain, chronic pain, musculoskeletal pain, post-operative pain, visceral pain, peripherally mediated pain, centrally mediated pain, inflammatory pain, neuropathic pain, nociceptive pain, and idiopathic pain.
  • the compounds, compositions and methods of the invention are of particular use in treating, preventing or lessening of pain includes but is not limited to acute pain, chronic pain, visceral pain, neuropathic pain, inflammatory pain or nociceptive pain or pain associated with, such as but are not limited to dental pain, eye pain, ear pain, perioperative, traumatic pain, muscle pain, pain in burned skin, sun burn, trigeminal neuralgia, spasm of the gastrointestinal tract or uterus, colics, back pain, chronic fatigue syndrome, clinical depression, complex regional pain syndrome, myofascial pain syndrome, post-vasectomy pain syndrome, restless leg syndrome, spinal stenosis, chronic pelvic pain, pancreatitis, peptic ulcer, interstitial cystitis, renal colic, angina, dysmenorrhoea, menstruation, gynaecological pain, irritable bowel syndrome (IBS), non- ulcer dyspepsia, non-cardiac chest pain, myocardial ischemia
  • the compounds of the invention may be useful for treating various types of inflammatory disease such as inflammations due to immune system, inflammations due to cancer, atherosclerosis, ischaemic heart diseases, pancreatitis, which includes but is not limited to acute pancreatitis and chronic pancreatitis, which is characterized by recurring or persistent abdominal pain with or without steatorrhea hereditary pancreatitis, pancreatic dysfunction.
  • inflammatory disease such as inflammations due to immune system, inflammations due to cancer, atherosclerosis, ischaemic heart diseases, pancreatitis, which includes but is not limited to acute pancreatitis and chronic pancreatitis, which is characterized by recurring or persistent abdominal pain with or without steatorrhea hereditary pancreatitis, pancreatic dysfunction.
  • Respiratory related syndromes, disorders or diseases include, but are not limited to, diseases of the respiratory tract or lung diseases such as asthma, bronchitis (acute or chronic), emphysema, allergic rhinitis, emphysema, adult respiratory distress syndrome (ARDS), pigeon fancier's disease, farmer's lung, chronic obstructive pulmonary disease (COPD), asthma including allergic asthma (atopic or non-atopic) as well as exercise- induced bronchoconstriction, occupational asthma, viral- or bacterial exacerbation of asthma, other non-allergic asthmas and "whez- infant syndrome", pneumoconiosis, including aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis.
  • diseases of the respiratory tract or lung diseases such as asthma, bronchitis (acute or chronic), emphys
  • the compounds of invention may be useful in the treatment of pruritus and related diseases including, but not limited to psoriatic pruritis, itch due to hemodyalisis, aguagenic pruritis, itching caused by skin disorders, allergic itch, insect bite itch, itch caused by hypersensitivity such as dry skin, acne, eczema, psoriasis or injury, itch caused by vulvar vestibulitis and the similar itch.
  • pruritus and related diseases including, but not limited to psoriatic pruritis, itch due to hemodyalisis, aguagenic pruritis, itching caused by skin disorders, allergic itch, insect bite itch, itch caused by hypersensitivity such as dry skin, acne, eczema, psoriasis or injury, itch caused by vulvar vestibulitis and the similar itch.
  • Allergic diseases all forms of allergic reactions include but are not limited to angioneurotic edema, hay fever, insect bites, viral or bacterial diseases, allergic reactions to drugs, blood derivatives, contrast agents, delayed or immediate hypersensitivity, allergic rhinitis, contact dermatitis, conjunctivitis, allergic reactions associated with inflammatory diseases such as diseases of the joints, spondylitis, gout, vasculitis, Crohn's disease, inflammatory bowel disease (IBD), irritable bowel syndrome (IBS) or osteoporosis.
  • inflammatory diseases such as diseases of the joints, spondylitis, gout, vasculitis, Crohn's disease, inflammatory bowel disease (IBD), irritable bowel syndrome (IBS) or osteoporosis.
  • Autoimmune or inflammation related syndromes, disorders or diseases include, but are not limited to, psoriasis, lupus erythematosus, diseases of the connective tissue, Sjogren's syndrome, ankylosing spondylarthritis, rheumatoid arthritis, reactional arthritis, undifferentiated spondylarthritis, Behcet's disease, autoimmune hemolytic anaemias, multiple sclerosis, amyotrophic lateral sclerosis, amyloses, graft rejection or diseases affecting the plasma cell line.
  • Gastrointestinal diseases including, but not limited to inflammatory bowel diseases, irritable bowel syndrome, regional enteritis (Crohns disease), colitis ulcerosa, gastritis, or aphthous.
  • the compounds described herein may be prepared by techniques known in the art.
  • the compounds described herein may be prepared by following the reaction sequence as depicted in Scheme-1 to Scheme-7.
  • specific bases, acids, reagents, solvents, coupling agents, etc. are mentioned, it is understood that other bases, acids, reagents, solvents, coupling agents etc., known in the art may also be used and are therefore included within the scope of the present invention.
  • Variations in reaction conditions, for example, temperature and/or duration of the reaction which may be used as known in the art are also within the scope of the present invention. All the isomers of the compounds in described in these schemes, unless otherwise specified, are also encompassed within the scope of this invention.
  • the compound of formula (12) wherein X, R, R 2 , R 3 , R 4 and 'm' are described herein above and is methyl or nitro, can be prepared by following the sequential transformations as depicted in Scheme-1 .
  • appropriately substituted benzaldehyde is reacted with malonic acid in presence of pyridine to give compound of formula (1 ).
  • the azide compound of formula (2) is prepared by treating acid compound of formula (1 ) with diphenylphosphoryl azide (DPPA) which is further converted to isoquinolinone of formula (3) by treating with iodine in suitable solvent.
  • Compound of formula (4) is prepared by treating compound formula (3) with bromine in presence of suitable solvent, and is further chlorinated with POCI 3 to give compound of formula (5).
  • This compound of formula (5) is converted to methoxy compound of formula (6) by reacting with sodium methoxide.
  • Compound of formula (8) is prepared from formula (6) by treating with n-butyl lithium followed by hydrolysis with sodium chlorite and disodium hydrogen phosphate in presence of suitable solvent.
  • the compound formula (1 0) is prepared by treating of compound of formula (8) with aqueous HBr followed by chlorination with POCI 3 in presence of suitable solvent.
  • This compound of formula (1 0) undergoes amide coupling reaction with formula R 2 -NH-R 3 in presence of HOBt (hydroxybenzotriazole), EDCI.HCI (1 -Ethyl-3-(3-dimethyl aminopropyl)carbodiimide Hydrochloride) and DIPEA (N,N- Diisopropylethylamine) in suitable solvent to afford compound formula (1 1 ).
  • HOBt hydroxybenzotriazole
  • EDCI.HCI (1 -Ethyl-3-(3-dimethyl aminopropyl)carbodiimide Hydrochloride
  • DIPEA N,N- Diisopropylethylamine
  • the compound of formula (12) wherein X, R, R 2 , R 3 , R 4 and 'm' are described herein above and is ethyl or cycloalkyl, can be prepared by following the sequential transformations as depicted in Scheme-2.
  • the compound of formula (15) can be obtained from optionally substituted isoquinoline compound by following three sequential transformations.
  • optionally substituted isoquinoline compound undergoes halogenation using NBS (N-bromosuccinimide) and dichloromethane followed by cyanation using palladium and Zn(CN) 2 to give cyano compound, which is further hydrolyzed to give compound of formula (15).
  • This acid compound is alkylated in presence of EDCI, HOBt and methanol to give compound of formula (16).
  • Compound of formula (1 6) is converted to halo compound of formula (1 7) in presence of NBS which again undergoes hydrolysis reaction to give acid compound of formula (18).
  • This acid compound is coupled with R 2 -NH-R 3 in presence of EDCI and HOBt and suitable solvent to obtain the compound of formula (1 9).
  • the compound of formula (20) is obtained by reacting a compound of formula (19) with R 1 B(OH) 2 in presence of palladium reagent.
  • the compound of formula (20) is oxidized to give N-oxide compound of formula (21 ) in presence of mCPBA which further halogenated with POCI 3 to give compound of formula (22).
  • this halo compound is converted to compound of formula (12) by reacting with R 4 -XH in presence of a ligand and palladium.
  • the compounds of formula (33), wherein R, R 2 , R 3 , R 4 , and 'm' are as described herein above, can be prepared by following the sequential transformations as depicted in Scheme-3.
  • Compound of formula (25) is prepared from compound of formula (24) by using suitable alkylating agent and base. Another hydroxyl group in compound of formula (25) is protected by using Tf 2 0 followed by alkylation using Ak-B(OH) 2 (where Ak is alkyl, cycloalkyi or haloalkyi etc.,) in presence of suitable reagent to afford compound of formula (27).
  • Compound of formula (28) is obtained from compound of formula (27) in presence of POCI 3 and DMF which is further converted to acid compound of formula (29).
  • the compound of formula (36) undergoes hydrolysis to give acid compound of formula (37) in the presence of aqueous potassium hydroxide (KOH) solution.
  • This acid compound of formula (37) further undergoes amide coupling reaction with an amine (R 2 -NH-R 3 ) in the presence of hydroxybenzotriazole (HOBt), 1 -ethyl-3-(3- dimethyllaminopropyl) carbodiimide hydrochloride (EDC.HCI) and N,N- diisopropylethylamine (DIPEA) to afford compound of formula (38).
  • HABt hydroxybenzotriazole
  • EDC.HCI 1 -ethyl-3-(3- dimethyllaminopropyl) carbodiimide hydrochloride
  • DIPEA N,N- diisopropylethylamine
  • the compounds of formula (44), where R, R 2 , R 3 , R 4 and 'm' are as described herein above, can be prepared by following the sequential transformations of compound formula (39) as depicted in Scheme-5.
  • the compound of formula (39) can be treated with S0 2 gas and N-Chlorosuccinimide (NCS) in presence of n-butyl Lithium to give compound of formula (40).
  • NCS N-Chlorosuccinimide
  • This compound of formula (40) can be reacted with amino derivative of formula (R 2 -NH-R 3 ) in presence of DIPEA to afford compound of formula (41 ) which undergoes hydrolysis to give compound of formula (42).
  • This hydroxyl compound of formula (42) can be treated with POCI 3 to give chloro compound of formula (43).
  • this chloro compound of formula (43) can be coupled with R 4 NH 2 to give compound of formula (44).
  • Compound of formula (12) wherein X, R, R 2 , R 3 , R 4 and 'm' are described herein above, can be prepared by following the sequential transformations as depicted in Scheme-6.
  • Compound of formula (45) is obtained where R-t is halogen, from compound of formula (16) by treating with N-halosuccinimide. Additionally, Compound of formula (45) is obtained where is hydroxy, cyano, amino, alkoxy, haloalkoxy or cycloalkoxy, from compound of formula (16) in presence of suitable reagent(s) and conditions.
  • lntermediate-2 o-Methylcinnamoyl azide:
  • lntermediate-3 5-Methylisoquinolin-1 (2A7)-one: A stirred solution of o-methylcinnamoyl azide (lntermediate-2) (1 0 g, 53.5 mmol) in toluene (100 mL) was heated to 1 10 2 C for 1 h. The solvent was removed, and charged 1 ,2-dichlorobenzene (DCB) (1 00 mL), l 2 (catalytic) and heated to 180 2 C and maintained for overnight. The solvent was removed under reduced pressure and the product was recrystallized (EtOAc:Hexanes; 90:10) to furnish the desired compound (4 g, 45%).
  • DCB ,2-dichlorobenzene
  • lntermediate-7 1 -Methoxy-5-methylisoquinolin-4-carbaldehyde:
  • lntermediate-8 1 -Methoxy-5-methylisoquinolin-4-carboxylic acid: To a stirred solution of 1 -methoxy-5-methylisoquinolin-4-carbaldehyde (lntermediate-7) (2 g, 1 0 mmol) in ACN-H 2 0 (120 mL, 1 :1 ) was added, NaCI0 2 (5.4 g, 59.7 mmol), NaH 2 P0 4 (5.5 g, 40 mmol) and the mixture was stirred at RT overnight. It was then extracted with EtOAc (3 x 150 mL), dried over Na 2 S0 4 , filtered and concentrated to give the desired acid derivative (1 .2 g, 56%).
  • the title compound was prepared by using 1 -chloro-5-methylisoquinolin-4- carboxylic acid (lntermediate-10) and (tetrahydro-2A7-pyran-4-yl)methanamine by following the similar procedure as described for lntermediate-1 1 a.
  • This compound was prepared by using 1 -chloro-5-methylisoquinolin-4-carboxylic acid (lntermediate-10) and ferf-butylamine by following the similar procedure as described for lntermediate-1 1 a.
  • This compound was prepared by using 1 -chloro-5-methylisoquinolin-4-carboxylic acid (lntermediate-10) and 2,6-dimethylpiperazine by following the similar procedure as described for intermediate-1 1 a.
  • lntermediate-1 1 i 2-Oxa-5-azabicyclo[2.2.1 ]heptan-3-yl(1 -chloro-5-methylisoquinolin-4-yl) methanone:
  • the title compound was prepared by using 1 -chloro-5-methylisoquinolin-4- carboxylic acid (lntermediate-10) and 2-phenylmorpholine by following the similar procedure as described for intermediate-1 1 a.
  • This compound was prepared by using 1 -chloro-5-methylisoquinolin-4-carboxylic acid (lntermediate-1 0) and cyclohexylamine by following the similar procedure as described for intermediate-1 1 a.
  • the title compound was prepared by using 1 -chloro-5-methylisoquinolin-4- carboxylic acid (lntermediate-1 0) and 1 ,1 -dioxidothiomorpholine by following the similar procedure as described for intermediate-1 1 a.
  • the title compound was prepared by using 1 -chloro-5-methylisoquinolin-4- carboxylic acid (lntermediate-10) and 4,4-difluoropiperidine by following the similar procedure as described for intermediate-1 1 a.
  • lntermediate-16 Methyl 5-bromoisoquinolin-4-carboxylate: To a mixture of cone. H 2 S0 4 (16 mL) and methyl isoquinoline-4-carboxylate (lntermediate-1 5) (2.8 g, 15 mmol) at 0 2 C was added, NBS (3.3 g, 18.7 mmol) slowly and the reaction mixture was allowed to come to RT and stirred overnight. After completion (TLC), it was quenched with ice, neutralized (NaHCC>3) and extracted with EtOAc (50 mL x 3).
  • 5-Alkyl-2-oxideisoquinolin-4-carboxylic amide In a 100 m L sealed tube, 5-alkylisoquinolin-4-carboxylic amide (lntermediate-1 9) (1 equiv) in CH 2 CI 2 (10 mL) was added, mCPBA (mefa-Chloroperoxybenzoic acid) (2 equiv) at 0 °C and the reaction mixture was stirred overnight while allowing it to attain RT. After completion (TLC), it was diluted with CH 2 CI 2 (50 mL) and washed with saturated NaHC0 3 solution. The organic phase was separated, dried and concentrated to give the corresponding N-oxide derivative (85-95%).
  • mCPBA mefa-Chloroperoxybenzoic acid
  • R is alkyl or cycloalkyl
  • R 2 and R 3 is as defined in Formula (I)
  • Step-1 5-Methoxynaphthalen-1 -ol:
  • Step-2 5-Methoxynaphthalen-1 -yl trifluoromethanesulfonate:
  • Step-3 1 -Methoxy-5-methyl naphthalene:
  • Step-4 4-Methoxy-8-methyl-1 -naphthaldehyde:
  • Step-5 4-Methoxy-8-methyl-1 -naphthoic acid:
  • Step-6 (4-Methoxy-8-methylnaphth lino)methanone:
  • Step-7 (4-Hydroxy-8-methylnaphth lino)methanone:
  • Step-8 5-Methyl-4-(morpholin-4-car -1 -yl trifluoromethanesulfonate:
  • Step-1 (4-Methoxy-8-methylnaphthalen-1 -yl)(piperidin-1 -yl)methanone:
  • Step-5 of lntermediate-22 was reacted with piperidine by following the similar procedure as described in Step-6 of lntermediate- 22.
  • This compound was prepared by using (4-methoxy-8-methylnaphthalen-1 - yl)(piperidin-1 -yl)methanone (above step-1 intermediate) through the similar method as described in Step-7 of lntermediate-22.
  • Step-3 5-Methyl-4-(piperidin-1 -carbonyl)naphthalen-1 -yl trifluoromethanesulfonate:
  • This compound was prepared by treating (4-hydroxy-8-methylnaphthalen-1 - yl)(piperidin-1 -yl)methanone (above step-2 intermediate) with Tf 2 0 by following the similar procedure as described in Step-8 of lntermediate-22
  • Step-1 (4-Methoxy-8-methylnaphthalen-1 -yl)(pyrrolidin-1 -yl)methanone:
  • Step-2 (4-Hydroxy-8-methylnaphthalen-1 -yl)(pyrrolidin-1 -yl)methanone:
  • This compound was prepared by using (4-methoxy-8-methylnaphthalen-1 - yl)(pyrrolidin-1 -yl)methanone (above step-1 intermediate) through the similar procedure as described in Step-7 of lntermediate-22.
  • Step-3 5-Methyl-4-(pyrrolidin-1 -carbonyl)naphthalen-1 -yl trifluoromethanesulfonate:
  • This compound was prepared by treating (4-hydroxy-8-methylnaphthalen-1 -yl)
  • Step-1 2-Oxa-5-azabicyclo[2.2.1 ]heptan-5-yl(4-methoxy-8-methylnaphthalen-1 -yl) methanone:
  • Step-2 2-Oxa-5-azabicyclo[2.2.1 ]heptan-5-yl(4-hydroxy-8-methylnaphthalen-1 -yl) methanone:
  • This compound was prepared by using 2-oxa-5-azabicyclo[2.2.1 ]heptan-5-yl(4- methoxy-8-methylnaphthalen-1 -yl)methanone (above step-1 intermediate) through the similar procedure as described in Step-7 of lntermediate-22.
  • Step-3 4-(2-Oxa-5-azabicyclo[2.2.1 ]heptan-5-carbonyl)-5-methylnaphthalen-1 -yl trifluoro methanesulfonate:
  • Step-1 3-Azabicyclo[3.1 .0]hexan-3-yl(4-methoxy-8-methylnaphthalen-1 -yl)methanone:
  • Step-2 3-Azabicyclo[3.1 .0]hexan-3-yl(4-hydroxy-8-methylnaphthalen-1 -yl)methanone: This compound was prepared by using 3-azabicyclo[3.1 .0]hexan-3-yl(4-methoxy- 8-methylnaphthalen-1 -yl)methanone (above step-1 intermediate) through the similar method as described in Step-7 of lntermediate-22.
  • Step-3 4-(3-Azabicyclo[3.1 .0]hexan-3-carbonyl)-5-methylnaphthalen-1 -yl trifluoromethane sulfonate:
  • the desired intermediate was prepared by treating 3-azabicyclo[3.1 .0]hexan-3- yl(4-hydroxy-8-methylnaphthalen-1 -yl)methanone (above step-2 intermediate) with Tf 2 0 using the similar procedure as described in Step-8 of lntermediate-22.
  • Step-1 6-Oxa-3-azabicyclo[3.1 .1 ]heptan-3-yl(4-methoxy-8-methylnaphthalen-1 - yl)methanone:
  • Step-2 6-Oxa-3-azabicyclo[3.1 .1 ]heptan-3-yl(4-hydroxy-8-methylnaphthalen-1 - yl)methanone:
  • This compound was prepared by using 6-oxa-3-azabicyclo[3.1 .1 ]heptan-3-yl(4- methoxy-8-methylnaphthalen-1 -yl)methanone (above step-1 intermediate) through the similar procedure as described in Step-7 of lntermediate-22.
  • Step-3 4-(6-Oxa-3-azabicyclo[3.1 .1 ]heptan-3-carbonyl)-5-methylnaphthalen-1 -yl trifluoro methanesulfonate:
  • the title compound was prepared by treating 6-oxa-3-azabicyclo[3.1 .1 ]heptan-3- yl(4-hydroxy-8-methylnaphthalen-1 -yl)methanone (above step-2 intermediate) with Tf 2 0 using the similar procedure as described in Step-8 of lntermediate-22.
  • Step-1 (4-Methoxy-8-methylnaphthalen-1 -yl)(2-oxa-6-azaspiro[3.3]heptan-6- yl)methanone:
  • Step-2 (4-Hydroxy-8-methylnaphthalen-1 -yl)(2-oxa-6-azaspiro[3.3]heptan-6- yl)methanone:
  • This compound was prepared by using (4-methoxy-8-methylnaphthalen-1 -yl)(2- oxa-6-azaspiro[3.3]heptan-6-yl)methanone (above step-1 intermediate) using similar procedure as described in Step-7 of lntermediate-22.
  • Step-3 5-Methyl-4-(2-oxa-6-azaspiro[3.3]heptan-6-carbonyl)naphthalen-1 -yl trifluoro methanesulfonate:
  • the title compound was prepared by treating (4-hydroxy-8-methylnaphthalen-1 - yl)(2-oxa-6-azaspiro[3.3]heptan-6-yl)methanone (above step-2 intermediate) with Tf 2 0 using the similar procedure as described in Step-8 of lntermediate-22.
  • Step-1 1 -Amino-4-cyanonaphthalene:
  • Step-3 4-((optionally substituted phenyl)amino)-1 -naphthoic acid:
  • Step-2 intermediate (1 equiv) in EtOH (40 imL) was added 50% aq. KOH solution (5 equiv) and the reaction mixture was refluxed for 1 day. The solvent was then removed under reduced pressure and to this, 1 N HCI solution was added drop wise (pH ⁇ 2) and extracted with EtOAc (3 x 200 imL), washed with water (50 imL), dried (Na 2 S0 4 ), filtered and concentrated to give the corresponding acid derivative of the title compound.
  • Step-1 5-Chloroisoquinolin-4-carboxylic acid
  • Step-2 5-Chloroisoquinolin-4-carboxylic amide
  • Step-3 5-Chloro-2-oxideisoquinolin-4-carboxylic amide
  • This intermediate was prepared from the corresponding morpholine amide derivative and POCI 3 by using the similar procedure as described above.
  • This intermediate was prepared from the corresponding morpholine derivative and POCI 3 by using the similar procedure as described for lntermediate-46.
  • This intermediate was prepared from the corresponding piperidine derivative and POCI 3 by using the similar procedure as described for lntermediate-46.
  • This compound was prepared by using lntermediate-11a and 4-fluoroaniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, CDCI 3 ) ⁇ 2.62 (s, 3H), 3.28-4.16 (m, 8H), 7.09-7.13 (m, 2H), 7.19 (s, 1 H), 7.51-7.61 (m, 4H), 7.86- 7.88 (m, 1 H), 7.95 (s, 1 H); MS m/z 366 (M+1 ).
  • This compound was prepared by using lntermediate-11 a and 3-chloro-2- fluoroaniline by following the similar procedure as described in Example-1 ; n H NMR (400 MHz, CDCI 3 ) ⁇ 2.61 (s, 3H), 3.27-4.14 (m, 8H), 7.05-7.12 (m, 2H), 7.48-7.49 (m, 1 H), 7.53-7.57 (m. 2H), 7.88-7.91 (m, 1 H), 7.99 (s. 1 H), 7.40-7.44 (m, 1 H); MS m/z 400 (M+1 ).
  • This compound was prepared by using lntermediate-11a and 3-chloro-4- fluoroaniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, CDCI 3 ) ⁇ 2.59 (s, 3H), 3.27-4.16 (m, 8H), 7.12-7.17 (s, 1 H, J - 8.8 Hz), 7.31 (s, 1H), 7.43-7.54 (m, 3H), 7.83-7.87 (m, 2H), 7.95 (s, 1 H); MS m/z 400 (M+1 ).
  • This compound was prepared by using lntermediate-11d and 3-
  • This compound was prepared by using lntermediate-11d and 2-chloro-4- fluorophenol by following the similar procedure as described in Example-54; H NMR (400 MHz, CDCW ⁇ 1.46-1.76 (m. 6H), 2.63 (s, 3H), 3.1 1-3.16 (m, 1 H), 3.33-3.50 (m, 2H), 4.09-4.14 (m, 1 H), 7.06-7.30 (m, 3H), 7.56-7.61 (m, 2H), 7.79 (s, 1 H), 8.43-8.45 (m, 1 H); MS m/z 399 (M+1 ).
  • the desired compound was prepared by using Intermediate-He and 3,5- difluoroaniline by following the similar procedure as described in Example-1; 1 H NMR (400 MHz. CDCI3) ⁇ (as mixture of isomers) 1.90-2.07 (m, 2H), 2.58 (s, 3H). 3.27-5.17 (m, 6H), 6.50-6.55 (m. 1H), 7.32-7.36 (m, 2H), 7.49-7.56 (m, 3H), 7.82-8.01 (m, 2H); MS m/z 396 (M+1 ).
  • the desired compound was prepared by using lntermediate-11 i and 3- (trif!uoromethyl) aniline by following the similar procedure as described in Example-1 ; H NMR (400 MHz, CDCI 3 ) ⁇ (as mixture of isomers) 1.85-2.04 (m, 2H), 2.60 (s, 3H), 3.23- 5.15 (m, 6H), 7.32-7.35 (m, 1 H), 7.44-7.56 (m, 4H), 7.83-8.02 (m, 4H); MS m/z 428 (M+1 ).
  • This compound was prepared by using lntermediate-11 i and 3-chloro-4- fluoroaniline by following the similar procedure as described in Example-1 ; H NMR (400 MHz, CDCIa) ⁇ (as mixture of isomers) 1.90-2.06 (m, 2H), 2.61 (s, 3H), 3.25-5. 5 (m, 6H), 7.13-7.19 (m, 1 H), 7.43-7.58 (m, 3H), 7.60-7.96 (m, 4H); MS m/z 412 (M+ ).
  • the desired compound was prepared by using lntermediate-11j and 3- (trifluoromethyl) aniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, CDCI 3 ) ⁇ 0.22-0.35 (m, 1 H), 0.77-0.83 (m, 1 H), 1.46-1.68 (m, 2H), 2.55 (s, 3H), 3.24-4.15 (m, 4H), 7.31 -7.33 (m, 1 H), 7.45-7.56 (m, 4H), 7.83-7.96 (m, 4H); MS m z 412 (M+1 ).
  • the desired compound was prepared by using lntermediate-11k and 3- (trifluoromethyl) aniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR (400 MHz, CDCI 3 ) ⁇ (as mixture of isomers) 1.78-2.1 (m, 4H), 2.58-2.68 (m, 2H), 2.76 (s, 3H), 3.60-4.09 (m, 6H), 4.83 (s, 1 H), 7.33-7.35 (d, 1 H, J 7.6 Hz), 7.42 (s, 1 H), 7.48-7.55 (m, 3H), 7.88-8.06 (m, 3H); MS m/z 456 (M+1).
  • the desired compound was prepared by using lntermediate-11 a and 3-fluoro-4- methylaniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, CDC ) ⁇ 2.24 (s, 3H), 2.57 (s, 3H), 3.25-4.12 (m, 8H), 7.12-7.14 (m, 2H), 7.23-7.24 (m, 1 H), 7.45-7.57 (m, 3H), 7.81-7.83 (m, 1 H), 7.93 (s, 1 H); MS m/z 380 (M+1 ).
  • the desired compound was prepared by using lntermediate-11 d and 3-chloro-4- methyl aniline by following the similar procedure as described in Example-1 ;
  • the desired compound was prepared by using lntermediate-11d and 4-fluoro-3- methyl aniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, CDCI 3 ) ⁇ 1.48-1.74 (m, 6H), 2.28 (s, 3H), 2.58 (s, 3H), 3.12-3.18 (m, 1 H), 3.40-3.48 (m, 2H), 4.1 1-4.15 (m, 1 H), 6.98-7.02 (m, 1 H), 7.31-7.52 (m, 5H), 7.82-7.84 (m, 1 H), 7.88 (s, 1 H); MS m/z 378 (M+1 ).
  • the desired compound was prepared by using lntermediate-11 n and 3,5- difluoroaniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, DMSO-d6) ⁇ 1 .77-1.98 (m, 4H), 2.45 (s, 3H), 2.99-3.57 (m, 4H), 6.77-6.81 (m, 1 H), 7.60-7.72 (m, 4H), 7.96 (s, 1 H), 8.41-8.44 (m, 1 H), 9.6 (s, 1 H); MS m/z 368 (M+1).
  • the desired compound was prepared by using lntermediate-1 1 n and 2,4- difluoroaniline by following the similar procedure as described in Example-1 ;
  • 1 H NMR 400 MHz, DMSO-d6) ⁇ 1.77-1.9 (m, 4H), 2.46 (s, 3H), 3.01-3.52 (m, 4H), 7.10-7.11 (m, 1 H), 7.29-7.35 (m, 1 H), 7.47-7.61 (m, 3H), 7.7 (s, 1 H), 8.32-8.35 (m, 1 H), 9.16 (s, 1 H); MS m/z 368 (M+1 ).
  • This compound was prepared by using lntermediate-11d and 3-chloro-2- methylaniline by following the similar procedure as described in Example-1 ; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 1.44-1.81 (m, 6 ⁇ ), 2.54 (s, 3 ⁇ ), 2.74 (s. 3 ⁇ ).
  • the desired compound was prepared by using lntermediate-1 1 d and 5-chloro-2- methylaniline by following the similar procedure as described in Example-1 ; 'H NMR (400 MHz, CDCI 3 ) ⁇ 1.51-1.78 (m. 6H), 2.33 (s, 3H), 2.62 (s. 3H), 3.15-3.21 (m, 1 H), 3.42-3.51 (m, 2H), 4.14-4.17 (m, 1 H), 7.0 (s.
  • the desired compound was prepared by using lntermediate-21a and 2,3- difluoroaniline by following the general procedure B; H NMR (400 MHz, CDCI 3 ) ⁇ 0.52- 0.55 (m, 1H), 0.88-0.97 (m, 2H), 1.02-1.03 (m, 1H), 2.46-2.48 (m, 1H), 3.3-3.34 (m, 1H), 3.39-3.42 (m, 1H), 3.54-3.63 (m, 2H), 3.7-3.77 (m, 2H), 3.93-3.96 (m, 1H), 4.32-4.35 (m, 1H), 6.88-6.92 (m, 1H), 7.11-7.15 (m, 1H), 7.46-7.47 (m, 1H), 7.56-7.62 (m, 2H), 7.91- 7.93 (dd, H), 8.07 (s, 1H).8.25-8.27 (m, 1H); MS m/z 410 (M+1).
  • This compound was prepared by using lntermediate-21 a and 2,3,6-trifluoroaniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 0.51 -0.54 (m, 1 ⁇ ), 0.87-0.96 (m, 2 ⁇ ), 1.01-1.02 (m, 1 H), 2.47 (m, 1 ⁇ ), 3.3-3.33 (m, 1 ⁇ ), 3.39-3.41 (m, 1 H), 3.5-3.62 (m, 2H), 3.7-3.91 (m, 2H), 3.91-3.94 (m,1 H), 4.29-4.32 (d, 1 H), 6.75 (bs,1 H), 6.95-7.08 (m, 2H), 7.53-7.63 (m, 2H), 7.98-7.99 (t, 2H); MS m/z 428 (M+1).
  • the desired compound was prepared by using lntermediate-21 a and 3- (trifluoromethyl) aniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 0.47-0.5 (m, 1 H), 0.85-0.94 (m, 2H), 0.97-1.01 (m, 1 H), 2.44-2.46 (m, 1 H), 3.29-3.35 (m, 1 H), 3.38-3.44 (m, 1 H), 3.58-3.66 (m, 2H), 3.71-3.78 (m, 2H), 3.93-3.96 (m,1 H), 4.32-4.35 (d, 1 H), 7.33-7.35 (d, 1 H), 7.47-7.56 (m, 4H), 7.86-7.91 (t, 2H), 7.98-8.01 (d, 2H); MS m/z 442 (M+1 ).
  • This compound was prepared by using lntermediate-21a and 2-fluoro-3- (trifluoromethyl) aniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 0.53-0.55 (m, 1H), 0.89-1.02 (m, 2H), 1.03-1.26 (m.1H), 2.43-2.5 (m, 1H), 3.27-3.33 (m, 1H), 3.34-3.44 (m, 1H), 3.53-3.64 (m, 2H), 3.7-3.74 (m, 2H), 3.93-3.96 (m, 1H), 4.32- 4.36 (d, 1H), 7.27-7.32 (m, 2H), 7.57-7.63 (m, 3H), 7.91-7.93 (d, 1H), 8.07 (s, 1H), 8.76- 8.8 (m, 1H); MS m/z 460 (M+1).
  • the desired compound was prepared by using lntermediate-21c and 2,3- difluoroaniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 1.29- 1.3 (t, 3H), 2.86-2.9 (m, 1H), 3.12-3.16 (m, 1H), 3.3-3.33 (m, 1H), 3.41-3.42 ((m, 1H), 3.6-3.67 (m, 3H), 3.7-3.71 (m, 1H), 3.79-3.82 (m, 1H), 4.11-4.12 (m, 1H), 6.91-6.93 (m, 1H), 7.1-7.16 (m.
  • This compound was prepared by using lntermediate-21d and 2,4-difIuoroaniline by following the general procedure B; H NMR (400 MHz. CDCI 3 ) ⁇ 1.32-1.36 (t, 3H), 1.49-1.52 (m, 2H), 1.61-1.78 (m, 4H), 2.85-2.91 (m, 1 H), 3.13-3.21 (m, 2H), 3.41 -3.52 (m, 2H), 4.15-4.18 (m, 1 H), 6.92-6.98 (m, 2H), 7.27-7.3 (m, 1 H), 7.58-7.64 (m, 2H), 7.88- 7.95 (m, 2H), 8.38-8.39 (m, 1 H); MS /z 396 (M+1).
  • the desired compound was prepared by using lntermediate-21d and 3,5- difluoroaniline by following the general procedure B; H NMR (400 MHz, CDCI 3 ) ⁇ 1.26- 1.31 (t, 3H), 1.52-1.56 (m, 2H), 1.59-1.81 (m, 4H), 2.81-2.87 (m, 1 H), 3.09-3.18 (m, 1 H), 3.2-3.23 (m, 1H), 3.46-3.54 (m, 2H), 4.18-4.22 (m, H), 6.49-6.52 (m, 1 H), 7.32-7.35 (dd, 2H), 7.5-7.57 (m, 2H), 7.62 (s, 1 H), 7.8-7.82 (m,1 H), 7.92 (s, 1 H); MS m/z 396 (M+1).
  • This compound was prepared by using lntermediate-21d and 3-chloroaniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 1.28-1.33 (t, 3H), 1.5- 1.54 (m, 2H), 1.62-1.79 (m, 4H), 2.82-2.88 (m, 1 H), 3.1-3.23 (m, 2H), 3.45-3.52 (m, 2H), 4.16-4.2 (m, 1 H), 7.03-7.05 (m, 1 H), 7.26-7.3 (m, 1 H), 7.42 (s, 1 H), 7.48-7.58 (m, 3H), 7.81-7.85 (m, 2H), 7.93 (s, 1 H); MS m/z 394 (M+1 ).
  • This compound was prepared by using lntermediate-21d and 3-chloro-2- fluoroaniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 1.33- 1.37 (t, 3H), 1.49-1.52 (m, 2H), 1.59-1.79 (m, 4H), 2.86-2.92 (m, 1 H), 3.14-3.2 (m, 2H), 3.4-3.53 (m, 2H), 4.14-4.19 (m, 1 H), 7.07-7.15 (m, 2H), 7.48 (brs, 1 H), 7.6-7.66 (m,2H), 7.9-7.92 (d, 1 H), 7.99 (s, 1 H), 8.14-8.45 (m, 1 H); MS m/z 412 (M+1 ).
  • This compound was prepared by using lntermediate-21 d and 2-fluoro-3- (trifluoromethyl) aniline by following the general procedure B; H NMR (400 MHz, CDCI 3 ) ⁇ 1.27-1.37 (t, 3H), 1.51-1.65 (m, 4H), 1.67-1.76 (m, 2H), 2.87-2.94 (m, 1 H), 3.15-3.21 (m, 2H), 3.41-3.53 (m, 2H), 4.16-4.19 (m, 1 H), 7.27-7.29 (m, 2H), 7.58-7.67 (m, 3H), 7.91-7.93 (d, 1 H), 8.0 (s, 1 H), 8.76-8.79 (m, 1 H); MS m/z 446 (M+1).
  • This compound was prepared by using lntermediate-21d and 3-fluoro-2- methylaniline by following the general procedure B; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 1.22- 1.36 (t, 3H), 1.48-1.51 (m, 2H), 1.59-1.77 (m, 4H), 2.24 (s, 3H), 2.87-2.91 (m, 1 H), 3.13- 3.19 (m, 2H), 3.44-3.52 (m, 2H), 4.12-4.16 (m, 1 H), 6.87-6.91 (t, 1H), 6.99 (br s, 1 H), 7.17 -7.23 (m, 1 H), 7.49-7.51 (d, H), 7.56-7.64 (m, 2H), 7.88-7.92 (m, 2H); MS m/z 392 (M+1 ).
  • This compound was prepared by using lntermediate-22 and m-toluidine using the similar procedure as described for Example-132; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 2.33 (s, 3H), 2.85 (s, 3H), 3.21-3.25 (m, 1 H), 3.28-3.3 (m, 1 H), 3.54-3.68 (m, 3H), 3.75-3.81 (m, 1 H), 3.88-3.92 (m, 1 H), 4.11-4.15 (m, 1 H), 6.80-6.82 (m.
  • the desired compound was prepared by using lntermediate-22 and 4-fluoro-3- methyl aniline using the similar procedure as described for Example-132; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 2.33 (s, 3 ⁇ ), 2.85 (s, 3 ⁇ ), 3.21-3.25 (m, 1H), 3.28-3.3 (m, 1 ⁇ ), 3.54-3.68 (m, 3H), 3.75-3.81 (m, 1 H), 3.88-3.92 (m, 1 H), 4.11 ⁇ .15 (m, 1 H), 6.80-6.82 (m.
  • the desired compound was prepared by using lntermediate-22 and 3-fluoro-4- methyl aniline using the similar procedure as described for Example-132; 1 H NMR (400 MHz, CDCI 3 ) ⁇ 2.42 (s, 3H), 2.68 (s, 3H), 3.22-3.30 (m, 2H), 3.57-3.6 (m.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pain & Pain Management (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Rheumatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

L'invention concerne des composés de Formule (I) ainsi que des procédés pour leur préparation qui sont utiles pour le traitement, la prévention et/ou la gestion des maladies, troubles, syndromes ou états associés à la modulation des récepteurs cannabinoïdes (CB). L'invention concerne également des méthodes de traitement, de prévention, de gestion et/ou de diminution des maladies, troubles, syndromes ou états associés à la modulation des récepteurs cannabinoïdes (CB) de Formule (I).
PCT/IB2011/056007 2010-12-30 2011-12-29 Modulateurs des récepteurs cannabinoïdes WO2012090177A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
IN226MUN2014 IN2014MN00226A (fr) 2011-07-05 2012-07-04
PCT/IB2012/053407 WO2013005168A2 (fr) 2011-07-05 2012-07-04 Modulateurs des récepteurs de cannabinoïdes
US13/542,234 US9006442B2 (en) 2011-07-05 2012-07-05 Cannabinoid receptor modulators

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
IN1477/KOL/2010 2010-12-30
IN1476KO2010 2010-12-30
IN1477KO2010 2010-12-30
IN1476/KOL/2010 2010-12-30
IN907/KOL/2011 2011-07-05
IN905/KOL/2011 2011-07-05
IN905KO2011 2011-07-05
IN908KO2011 2011-07-05
IN908/KOL/2011 2011-07-05
IN907KO2011 2011-07-05

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/542,234 Continuation-In-Part US9006442B2 (en) 2011-07-05 2012-07-05 Cannabinoid receptor modulators

Publications (2)

Publication Number Publication Date
WO2012090177A2 true WO2012090177A2 (fr) 2012-07-05
WO2012090177A3 WO2012090177A3 (fr) 2012-08-23

Family

ID=45531493

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2011/056007 WO2012090177A2 (fr) 2010-12-30 2011-12-29 Modulateurs des récepteurs cannabinoïdes

Country Status (1)

Country Link
WO (1) WO2012090177A2 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013005168A3 (fr) * 2011-07-05 2013-04-25 Lupin Limited Modulateurs des récepteurs de cannabinoïdes
JP2016510000A (ja) * 2013-02-20 2016-04-04 カラ ファーマシューティカルズ インコーポレイテッド 治療用化合物およびその使用

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002042248A2 (fr) 2000-11-24 2002-05-30 Novartis Ag Derives de naphtalene
WO2006129178A1 (fr) 2005-06-02 2006-12-07 Glenmark Pharmaceuticals S.A. Nouveaux ligands des recepteurs des cannabinoides, compositions pharmaceutiques contenant ces ligands, et procede servant a leur preparation
WO2007091950A1 (fr) 2006-02-07 2007-08-16 Astrazeneca Ab Benzimidazoles et imidazopyridines utiles dans le traitement de maladies ou de troubles associés au récepteur cannabinoïde 2 (cb2) tels que la douleur
WO2007102059A1 (fr) 2006-03-06 2007-09-13 Pfizer Japan Inc. Derives de sulfonyl benzimidazole
WO2008027812A2 (fr) 2006-08-28 2008-03-06 Forest Laboratories Holdings Limited Dérivés d'imidazopyridine et d'imidazopyrimidine
WO2008115672A1 (fr) 2007-03-21 2008-09-25 Janssen Pharmaceutica N.V. Procédé de traitement d'une douleur induite par un récepteur cb2
WO2009053799A1 (fr) 2007-10-24 2009-04-30 Glenmark Pharmaceuticals, S.A. Nouveaux ligands du récepteur cannobinoïde, compositions pharmaceutiques les contenant et leur procédé de préparation
WO2010084767A1 (fr) 2009-01-22 2010-07-29 Raqualia Pharma Inc. Composés de sulfone hétérocycliques saturés n-substitués ayant une activité agoniste du récepteur cb2
WO2010096371A2 (fr) 2009-02-18 2010-08-26 Boehringer Ingelheim International Gmbh Composés hétérocycliques qui modulent le récepteur cb2
WO2010133973A1 (fr) 2009-05-20 2010-11-25 Universite Lille 2 Droit Et Sante Dérivés de la 1,4-dihydropyridine et leurs utilisations

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE0103325D0 (sv) * 2001-10-04 2001-10-04 Astrazeneca Ab Novel compounds
WO2006038594A1 (fr) * 2004-10-04 2006-04-13 Ono Pharmaceutical Co., Ltd. Inhibiteur de canal de calcium de type n
US7812040B2 (en) * 2005-06-22 2010-10-12 Pfizer Inc. Histamine-3 receptor antagonists
EP1957073B1 (fr) * 2005-12-05 2014-04-23 Otsuka Pharmaceutical Co., Ltd. Substance medicamenteuse
WO2008121570A1 (fr) * 2007-03-29 2008-10-09 Irm Llc Composés et procédés pour moduler des récepteurs couplés à la protéine g
EP2152671A1 (fr) * 2007-05-22 2010-02-17 Prosidion Limited Composés d'aryle et d'hétéroaryle bicycliques pour le traitement de troubles métaboliques
EP2173743A2 (fr) * 2007-07-13 2010-04-14 Icagen, Inc. Inhibiteurs des canaux sodiques

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002042248A2 (fr) 2000-11-24 2002-05-30 Novartis Ag Derives de naphtalene
WO2006129178A1 (fr) 2005-06-02 2006-12-07 Glenmark Pharmaceuticals S.A. Nouveaux ligands des recepteurs des cannabinoides, compositions pharmaceutiques contenant ces ligands, et procede servant a leur preparation
WO2007091950A1 (fr) 2006-02-07 2007-08-16 Astrazeneca Ab Benzimidazoles et imidazopyridines utiles dans le traitement de maladies ou de troubles associés au récepteur cannabinoïde 2 (cb2) tels que la douleur
WO2007102059A1 (fr) 2006-03-06 2007-09-13 Pfizer Japan Inc. Derives de sulfonyl benzimidazole
WO2008027812A2 (fr) 2006-08-28 2008-03-06 Forest Laboratories Holdings Limited Dérivés d'imidazopyridine et d'imidazopyrimidine
WO2008115672A1 (fr) 2007-03-21 2008-09-25 Janssen Pharmaceutica N.V. Procédé de traitement d'une douleur induite par un récepteur cb2
WO2009053799A1 (fr) 2007-10-24 2009-04-30 Glenmark Pharmaceuticals, S.A. Nouveaux ligands du récepteur cannobinoïde, compositions pharmaceutiques les contenant et leur procédé de préparation
WO2010084767A1 (fr) 2009-01-22 2010-07-29 Raqualia Pharma Inc. Composés de sulfone hétérocycliques saturés n-substitués ayant une activité agoniste du récepteur cb2
WO2010096371A2 (fr) 2009-02-18 2010-08-26 Boehringer Ingelheim International Gmbh Composés hétérocycliques qui modulent le récepteur cb2
WO2010133973A1 (fr) 2009-05-20 2010-11-25 Universite Lille 2 Droit Et Sante Dérivés de la 1,4-dihydropyridine et leurs utilisations

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
ALSO, BIOORGANIC MEDICINAL CHEMISTRY, vol. 19, 2011, pages 939 - 950
ANAND ET AL., PAIN, vol. 138, 2009, pages 667 - 680
BELTRAMO ET AL., EUR. J. NEUROSCI., vol. 23, 2006, pages 1530 - 1538
BENITO ET AL., J NEUROSCI., vol. 23, 2003, pages 11136 - 11141
CABRAL ET AL., BR. J. PHARMACOL., vol. 153, 2008, pages 240 - 251
CABRAL ET AL.: "Handb. Exp. Pharmacol.", vol. 168, 2005, pages: 385 - 423
CARLISLE ET AL., INT. IMMUNO PHARMACOL., vol. 2, 2002, pages 69 - 82
DITTEL B N. ET AL., BJP, vol. 153, no. 2, 2008, pages 271 - 276
DUNCAN M MOUIHATE ET AL., AM. J. PHYSIOL. GASTROINTEST. LIVER PHYSIOL., vol. 295, pages G78 - G87
KLEGERIS ET AL., BR. J. PHARMACOL, vol. 139, 2003, pages 775 - 786
MARESZ ET AL., NAT. MED., vol. 13, 2007, pages 492 - 497
MARESZ K. ET AL., NATURE MEDICINE., vol. 13, 2007, pages 492 - 497
MOLINA-HOLGADO ET AL., EUR. J. NEUROSCI., vol. 25, 2007, pages 629 - 634
MUNRO ET AL., NATURE, vol. 365, 1993, pages 61 - 65
OFEK ET AL., PNAS, vol. 103, 2006, pages 696 - 701
PALAZUELOS J. ET AL., FASEB J., vol. 20, 2006, pages 2405 - 2407
PERTWEE ET AL.: "Handb. Exp. Pharmacol.", vol. 168, 2005, pages: 1 - 51
PHARMACOLOGY, BIOCHEMISTRY AND BEHAVIOR, vol. 90, 2008, pages 501 - 511
PIETR ET AL., FEBS LETT., vol. 583, 2009, pages 2071 - 2076
STELLA ET AL., GLIA, vol. 48, 2004, pages 267 - 277
WALTER ET AL., J. NEUROSCI., vol. 23, 2003, pages 1398 - 1405
YIANGOU ET AL., BMC. NEUROL., vol. 6, 2006, pages 12
ZHANG ET AL., EUR. J. NEUROSCI., vol. 17, 2003, pages 2750 - 2754

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013005168A3 (fr) * 2011-07-05 2013-04-25 Lupin Limited Modulateurs des récepteurs de cannabinoïdes
JP2016510000A (ja) * 2013-02-20 2016-04-04 カラ ファーマシューティカルズ インコーポレイテッド 治療用化合物およびその使用
JP2021006574A (ja) * 2013-02-20 2021-01-21 カラ ファーマシューティカルズ インコーポレイテッド 治療用化合物およびその使用

Also Published As

Publication number Publication date
WO2012090177A3 (fr) 2012-08-23

Similar Documents

Publication Publication Date Title
AU2017294231B2 (en) Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof
JP6689805B2 (ja) カテプシンcの置換2−アザ−ビシクロ[2.2.1]ヘプタン−3−カルボン酸(ベンジル−シアノ−メチル)−アミド阻害剤
US10233152B2 (en) LSD1 inhibitors
US7842703B2 (en) Substituted benzofused derivatives and their use as vanilloid receptor ligands
JP5608655B2 (ja) P2x3受容体活性のモジュレーター
US10336697B2 (en) Spiro[cyclobutane-1,3′-indolin]-2′-one derivatives as bromodomain inhibitors
CN106793775B (zh) 二氢碟啶酮衍生物及其用途
EA021367B1 (ru) Производное пиридин-3-карбоксамида
US11802126B2 (en) Imidazopyridinyl compounds and use thereof for treatment of neurodegenerative disorders
WO2009010824A1 (fr) Ligands des récepteurs vanilloïdes
CA3066328A1 (fr) Composes chimiques utilises comme inhibiteurs de la voie atf4
US9006442B2 (en) Cannabinoid receptor modulators
US11827640B2 (en) Substituted pyrazolo[1,5-a]pyrimidines as CFTR modulators
WO2012090177A2 (fr) Modulateurs des récepteurs cannabinoïdes
SG172924A1 (en) Dipeptidyl peptidase iv inhibitors
WO2012090179A2 (fr) Dérivés d'isoquinoline en tant que modulateurs des récepteurs cannabinoïdes
CN111205244B (zh) 噻唑并环类化合物、其制备方法、中间体和应用
JP2024516194A (ja) Pd1/pd-l1阻害剤としての化合物及びその方法
WO2021113368A1 (fr) Antagonistes de sstr5
TW202023548A (zh) 新穎噻唑衍生物以及其藥用可接受鹽類
EP2917208A1 (fr) Composés [3-hétéroaryl-2-trifluorométhyl-propyl]-pipéridin-1-yle ou -morpholin-4-yle en tant qu'antagonistes de trpa1 pour le traitement de maladies respiratoires
TW201329060A (zh) 做為trpv3調節劑的色原烷衍生物
BRPI0617995B1 (pt) Composto

Legal Events

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

Ref document number: 11813450

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11813450

Country of ref document: EP

Kind code of ref document: A2