WO2007021308A1 - Metabotropic glutamate-receptor-potentiating isoindolones - Google Patents

Metabotropic glutamate-receptor-potentiating isoindolones Download PDF

Info

Publication number
WO2007021308A1
WO2007021308A1 PCT/US2006/005246 US2006005246W WO2007021308A1 WO 2007021308 A1 WO2007021308 A1 WO 2007021308A1 US 2006005246 W US2006005246 W US 2006005246W WO 2007021308 A1 WO2007021308 A1 WO 2007021308A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
alkylnr
cycloalkyl
alkylaryl
Prior art date
Application number
PCT/US2006/005246
Other languages
French (fr)
Inventor
Bradford Van Wagenen
Radhakrishnan Ukkiramapandian
Joshua Clayton
Ian Egle
James Empfield
Methvin Isaac
Fupeng Ma
Abdelmalik Slassi
Gary Steelman
Rebecca Urbanek
Sally Walsh
Original Assignee
Astrazeneca Ab
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
Priority claimed from PCT/US2005/028760 external-priority patent/WO2006020879A1/en
Application filed by Astrazeneca Ab filed Critical Astrazeneca Ab
Priority to JP2008525976A priority Critical patent/JP5031745B2/en
Priority to US12/063,007 priority patent/US7807706B2/en
Priority to EP06720758A priority patent/EP1912939A1/en
Publication of WO2007021308A1 publication Critical patent/WO2007021308A1/en
Priority to US12/861,336 priority patent/US8153638B2/en

Links

Classifications

    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • C07D209/46Iso-indoles; Hydrogenated iso-indoles with an oxygen atom in position 1
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates to novel compounds that function as potentiators of glutamate receptors, methods for their preparation, pharmaceutical compositions containing them and their use in therapy.
  • the metabotropic glutamate receptors are a family of GTP -binding-protein (G-protein) coupled receptors that are activated by glutamate, and that have important roles in synaptic activity in the central nervous system, neural plasticity, neural development and neurodegeneration.
  • Activation of mGluRs in intact mammalian neurons elicits one or more of the following responses: activation of phospholipase C; increases in phosphoinositide (PI) hydrolysis; intracellular calcium release; activation of phospholipase D; activation or inhibition of adenyl cyclase; increases or decreases in the formation of cyclic adenosine monophosphate (cAMP); activation of guanylyl cyclase; increases in the formation of cyclic guanosine monophosphate (cGMP); activation of phospholipase A 2 ; increases in arachidonic acid release; and increases or decreases in the activity of voltage- and ligand-gated ion channels (Schoepp et al, 1993, Trends Pharmacol.
  • PI phosphoinositide
  • Group-I includes mGluRl and mGluR5, which activate phospholipase C and the generation of an intracellular calcium signal.
  • Group-II mGluR2 and niGluR3
  • Group-Ill mGluR4, rnGluR ⁇ , mGluR7, and mGluR8
  • mGluRs mediate an inhibition of adenylyl cyclase activity and cyclic AMP levels.
  • mGluR family receptors Activity of mGluR family receptors is implicated in a number of normal processes in the mammalian CNS, and are important targets for compounds for the treatment of a variety of neurological and psychiatric disorders. Activation of mGluRs is required for induction of hippocampal long-term potentiation and cerebellar long-term depression (Bashir et ah, 1993, Nature, 363:347 ; Bortolotto et a!., 1994, Nature, 368:740 ; Aiba et al., 1994, Cell, 79:365 ; Aiba et ah, 1994, Cell, 79:377).
  • mGluR activation has been suggested to play a modulatory role in a variety of other normal processes including synaptic transmission, neuronal development, apoptotic neuronal death, synaptic plasticity, spatial learning, olfactory memory, central control of cardiac activity, waking, motor control and control of the vestibulo-ocular reflex (Nakanishi, 1994, Neuron, 13:1031; Pin et ah, 1995, Neuropharmacology, see above; Knopfel et al., 1995, J. Med. Chem., 38:1417).
  • R 1 is a 3- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said ring is substituted by one or more B;
  • R 2 and R 3 are independently selected from the group consisting of H, Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, heterocycloalkyl, Cs.g-cycloalkyl, Ci -6 -alkyl-aryl, Ci -6 -alkyl-heteroaryl, and Ci- ⁇ -alkyl-Cs-s-cycloalkyl, wherein R and R 3 may be substituted by one or more A;
  • R 4 and R 6 are independently selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, Ci -6 -alkyl, Q-e-alkylhalo, OCi -6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2 - 6 -alkenyl, C 2 - 6 -alkynyl, OC 2-6 -alkynyl, C ⁇ s-cycloalkyl, Ci -6 -alkyl-C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3-8 ⁇ cycloalkyl, aryl, C 1-6 -alkylaryl, OC 0 .
  • 6 -alkylCO 2 R 10 d-e-alkylcyano, OC 2- 6-alkylcyano, C 0-6 -alkylNR 10 R ⁇ , OC 2 - 6 -alkylNR 10 R ⁇ , C 1 . 6 -alkyl(CO)NR 10 R 11 , OC 1-6 -alkyl(CO)NR 10 R 11 , C 0-6 -alkylNR 10 (CO)R n , OC 2-6 -alkylNR 10 (CO)R 11 , Co -6 -alkylNR 10 (CO)NR 10 R ⁇ , C 0 .
  • R 5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci_ 6 -alkyl, OCo -6 -alkyl, Ci_ 6 -alkylhalo, OC 1-6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2 - 6 -alkynyl, C 3 _ 8 -cycloalkyl, Ci-e-alkyl-Cs-s-cycloalkyl, OC 0-6 -alkyl-C 3-8 -cycloalkyl, aryl, Ci- 6 -alkylaryl, C 1-6 -alkylheteroaryl, OCi -6 -alkylaryl, OC 1-6 -alkylheteroaryl, Ci- ⁇ -alkylheterocycloalkyl, Oheterocycloalkyl, OQ
  • R 7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi -4 -alkyl, Ci -6 -alkyl, C 1-6 -alkylhalo, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, and C 3-8 -cycloalkyl;
  • R 8 and R 9 are independently selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, C 1-6 -alkyl, C 1-6 -alkylhalo, OC 1-6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl,
  • R 1 and R l are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, C 1-6 -alkyl, Ci -6 -alkylhalo, OCi -6 alkyl, OCi- 6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -cycloalkyl, Ci -6 -alkyl-C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3-8 -cycloalkyl, aryl, C 1-6 -alkylaryl, OC 0-6 -alkylaryl, Co- ⁇ -alkyl-heterocycloalkyl, OCj- ⁇ -alkyl-heterocycloalkyl, heteroaryl, and Ci -6 alkylheter
  • A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, Ci -6 -alkyl, Ci -6 -alkylhalo, OC 1-6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -cycloalkyl, C 1-6 -alkyl-C 3-8 -cycloalkyl, OCo- ⁇ -alkyl-Cs-s-cycloalkyl, aryl, Ci -6 -alkylaryl, OCo -6 -alkylaryl, d- ⁇ -alkyl-heterocyclyl, Ci-e-alkyl-heterocycloalkyl, OC 0-6 -alkyl-heterocycloalkyl, (CO
  • B is selected from the group consisting of C 3-8 -cycloalkyl, C 1-6 -alkyl-C 3- g-cycloalkyl, OCo- ⁇ -alkyl-C B -s-cycloalkyl, C 0-6 -alkylaryl, OC 0-6 -alkylaryl, Ci- ⁇ -alkyl-heterocycloalkyl, C 1-6 -alkyl-heterocycloalkyl, OCo- ⁇ -alkyl-heterocycloalkyl, C 0-6 -alkyl-heteroaryl and OC 0-6 -alkyl-heteroaryl, wherein any cyclic moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO 2 R, SO 2 R and CN; and n is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7,
  • the invention also provides processes for the preparation of compounds of formula I.
  • the invention further provides a pharmaceutical composition comprising a compound according to formula I together with a pharmaceutically acceptable carrier or excipient; in another aspect the invention provides a method for the treatment or prevention of neurological and psychiatric disorders associated with glutamate dysfunction in an animal in need of such treatment.
  • the method comprises the step of administering to the animal a therapeutically effective amount of a compound of formula I or a pharmaceutical composition thereof.
  • the invention also provides for the use of a compound according to formula I, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for the treatment of any of the conditions discussed herein. Further, the invention provides a compound of formula I, or a pharmaceutically acceptable salt or solvate thereof, for use in therapy.
  • the present invention is based upon the discovery of compounds that exhibit activity as modulators of metabotropic glutamate receptors. More particularly, the compounds of the present invention exhibit activity as potentiators of the mGluR2 receptor, and are useful in therapy, in particular as pharmaceuticals for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction.
  • alkyl as used herein means a straight- or branched-chain hydrocarbon radical having, for example, from one to six carbon atoms, and includes methyl, ethyl, propyl, isopropyl, t-butyl and the like.
  • alkenyl as used herein means a straight- or branched-chain alkenyl radical having, for example, from two to six carbon atoms, and includes ethenyl, 1 -propenyl, 1-butenyl and the like.
  • alkynyl as used herein means a straight- or branched-chain alkynyl radical having, for example, from two to six carbon atoms, and includes 1-propynyl (propargyl), 1-butynyl and the like.
  • cycloalkyl as used herein means a cyclic group (which may be unsaturated) having, for example, from three to seven carbon atoms, and includes cyclopropyl, cyclohexyl, cyclohexenyl and the like.
  • heterocycloalkyl as used herein means, for example, a three- to seven-membered cyclic group (which may be unsaturated) having at least one heteroatom selected from the group consisting of N, S and O, and includes piperidinyl, piperazinyl, pyrrolidinyl, tetrahydrofuranyl and the like.
  • alkoxy as used herein means a straight- or branched-chain alkoxy radical having, for example, from one to six carbon atoms and includes methoxy, ethoxy, propyloxy, isopropyloxy, t-butoxy and the like.
  • halo as used herein means halogen and includes fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms.
  • aryl as used herein means an aromatic group having, for example, five to twelve atoms, and includes phenyl, naphthyl and the like.
  • heteroaryl means an aromatic group which includes at least one heteroatom selected from the group consisting of N, S and O, and includes groups and includes pyridyl, indolyl, furyl, benzofuryl, thienyl, benzothienyl, quinolyl, oxazolyl and the like.
  • alkanoyl as used herein means a straight- or branched-chain alkanoyl radical having, for example, from two to seven atoms, and includes acetyl, propionyl, butyryl and the like.
  • cycloalkenyl as used herein means an unsaturated cylcloaklyl group having, for example, from four to seven carbon atoms, and includes cyclopent-1-enyl, cyclohex-1-enyl and the like.
  • alkylaryl refers to an alkyl radical substituted with an aryl, heteroaryl or cycloalkyl group, and includes 2-phenethyl, 3 -cyclohexyl propyl and the like.
  • the term "5- to 7-membered ring that may contain one or more heteroatoms independently selected from N, O and S” includes aromatic and heteroaromatic rings, as well as carbocyclic and heterocyclic rings which may be saturated or unsaturated, and includes furyl, isoxazolyl, oxazolyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, thiazolyl, thienyl, imidazolyl, triazolyl, morpholinyl, piperazinyl, piperidinyl, homopiperidinyl, tetrahydropyranyl, phenyl, cyclohexyl, cycloheptyl, cyclopentyl, cyclohexanyl and the like.
  • pharmaceutically acceptable salt means either an acid addition salt or a basic addition salt which is compatible with the treatment of patients.
  • a "pharmaceutically acceptable acid addition salt” is any non-toxic organic or inorganic acid addition salt of the base compounds represented by Formula I or any of its intermediates.
  • Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate.
  • Illustrative organic acids which form suitable salts include the mono-, di- and tricarboxylic acids.
  • Illustrative of such acids are, for example, acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, salicylic, 2-phenoxybenzoic, p-toluenesulfonic acid and other sulfonic acids such as methanesulfonic acid and 2-hydroxyethanesulfonic acid.
  • Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated, solvated or substantially anhydrous form.
  • the acid addition salts of these compounds are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • the selection criteria for the appropriate salt will be known to one skilled in the art.
  • Other non-pharmaceutically acceptable salts e.g. oxalates may be used for example in the isolation of compounds of Formula I for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • a "pharmaceutically acceptable basic addition salt” is any non-toxic organic or inorganic base addition salt of the acid compounds represented by Formula I or any of its intermediates.
  • Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxides.
  • Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethyl amine and picoline or ammonia.
  • the selection of the appropriate salt may be important so that an ester functionality, if any, elsewhere in the molecule is not hydrolyzed. The selection criteria for the appropriate salt will be known to one skilled in the art.
  • Solvate means a compound of Formula I or the pharmaceutically acceptable salt of a compound of Formula I wherein molecules of a suitable solvent are incorporated in a crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered as the solvate. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a hydrate.
  • stereoisomers is a general term for all isomers of the individual molecules that differ only in the orientation of their atoms in space. It includes mirror image isomers (enantiomers), geometric (cis/trans) isomers and isomers of compounds with more than one chiral centre that are not mirror images of one another (diastereomers).
  • treat or “treating” means to alleviate symptoms, eliminate the causation of the symptoms either on a temporary or permanent basis, or to prevent or slow the appearance of symptoms of the named disorder or condition.
  • terapéuticaally effective amount means an amount of the compound which is effective in treating the named disorder or condition.
  • pharmaceutically acceptable carrier means a non-toxic solvent, dispersant, excipient, adjuvant or other material which is mixed with the active ingredient in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to the patient.
  • a pharmaceutical composition i.e., a dosage form capable of administration to the patient.
  • a pharmaceutically acceptable oil typically used for parenteral administration.
  • R 1 is a 3- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said ring is substituted by one or more B;
  • R 2 and R 3 are independently selected from the group consisting of H, Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, heterocycloalkyl, C 3-8 -cycloalkyl, Ci -6 -alkyl-aryl, Ci_ 6 -alkyl-heteroaryl, Ci_ 6 -alkyl-heterocycloalkyl, and Ci -6 -alkyl-C 3-8 -cycloalkyl, wherein R 2 and R 3 may be substituted by one or more A;
  • R 4 and R 6 are independently selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, C 1-6 -alkyl, Ci -6 -alkylhalo, OC 1-6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC2-6-alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -Cy cloalkyl, Ci- ⁇ -alkyl-Cs.s-cycloalkyl, OCo -6 -alkyl-C 3-8 -cycloalkyl, aryl, C 1-6 -alkylaryl, OC 0-6 -alkylaryl, (CO)R 10 , O(CO)R 10 , 0(CO)OR 10 , C(O)OR 10 , O(CNR 10 )OR n , C 1-6 -alkylOR 10
  • R 5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci -6 -alkyl, OCo -6 -alkyl, C 1-6 -alkylhalo, OC 1-6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3 .g-cycloalkyl, Ci- ⁇ -alkyl-Cs-s-cycloalkyl, OCo-o-alkyl-Cs-s-cycloalkyl, aryl, Cj -6 -alkylaryl, C 1-6 -alkylheteroaryl, OC 1-6 -alkylaryl, OC 1-6 -alkylheteroaryl, Ci- ⁇ -alkylheterocycloalkyl, Oheterocycloalkyl, OC ⁇ -alkylhe
  • R 7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi -4 -alkyl, Ci- 6 -alkyl, C 1-6 -alkylhalo, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, and C 3 -s-cycloalkyl; o Q
  • R and R are independently selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, C 1-6 -alkyl, C 1-6 -alkylhalo, OC 1-6 alkyl, OC 1-6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, and OC 2-6 -alkynyl, or, where n is greater than 1 , two or more R and/or R 9 on adjacent carbon atoms may be absent to form an alkenyl or alkynyl moiety;
  • R 10 and R 11 are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, C 1-6 -alkyl, Ci -6 -alkylhalo, OC 1-6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -cycloalkyl, Ci -6 -alkyl-C 3- g-cycloalkyl, OCo- 6 -alkyl-Cs-s-cycloalkyl, aryl, Ci- 6 -alkylaryl, OC 0-6 -alkylaryl, Co-e-alkyl-heterocycloalkyl, OCi- ⁇ -alkyl-heterocycloalkyl, heteroaryl, and Ci -6 alkylheteroary
  • A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, C 1-6 -alkyl, C 1-6 -alkylhalo, OCi -6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -cycloalkyl, C 1-6 -alkyl-C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3-8 -cycloalkyl, aryl, C 1-6 -alkylaryl, OC 0-6 -alkylaryl, Cj-e-alkyl-heterocyclyl, Ci-e-alkyl-heterocycloalkyl, OCo-e-alkyl-heterocycloalkyl, (CO)
  • B is selected from the group consisting of C ⁇ s-cycloalkyl, Ci -6 -alkyl-C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3- 8-cycloalkyl, C 0-6 -alkylaryl, OC 0-6 -alkylaryl, Ci-e-alkyl-heterocycloalkyl, OCo- ⁇ -alkyl-heterocycloalkyl, C 0-6 -alkyl-heteroaryl and OCo -6 -alkyl-heteroaryl, wherein any cyclic moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO 2 R, SO 2 R and CN, and n is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8; or a pharmaceutically acceptable salt, hydrate, solvate, optical is
  • R 1 is phenyl wherein said phenyl is substituted by one or more B;
  • R 2 and R 3 are independently selected from the group consisting of H, Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, heterocycloalkyl, C 3-8 -cycloalkyl, Ci -6 -alkyl-aryl, C 1-6 -alkyl-heteroaryl, Ci-e-alkyl-heterocycloalkyl, and C 1-6 -alkyl-C 3-8 -cycloalkyl, wherein R 2 and R 3 may be substituted by one or more A; R 4 and R 6 are independently selected from the group consisting of H 5 hydroxy, F, Cl, Br, I, nitro, cyano, Ci -6 -alkyl, Ci -6 -alkylhalo, OCi.
  • R 5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci -6 -alkyl, OCo- 6 -alkyl, Ci- 6 -alkylhalo, OC 1-6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3-8 -cycloalkyl, aryl, C 1-6 -alkylaryl, Ci -6 -alkylheteroaryl, OC 1-6 -alkylaryl, OC 1-6 -alkylheteroaryl, Ci- ⁇ -alkylheterocycloalkyl, Oheterocycloalkyl, OCi-e-alkylheterocycloalkyl, C(O)H, (CO)R
  • R 7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi -4 -alkyl, Ci -6 -alkyl, Ci -6 -alkylhalo, OC 1-6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, and C 3-8 -cycloalkyl;
  • R 8 and R 9 are both H
  • R 10 and R 11 are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, Ci -6 -alkyl, Ci_ 6 -alkylhalo, OCi -6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3-8 -cycloalkyl, Ci.
  • any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S and any cyclic moiety is optionally substituted with a substituent selected from alkyl, halo, hydroxyl, Oalkyl, haloalkyl and Ohaloalkyl;
  • A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, Ci -6 -alkyl, C 1-6 -alkylhalo, OC 1-6 alkyl, OCi -6 -alkylhalo, C 2-6 -alkenyl, OC 2-6 -alkenyl, C 2-6 -alkynyl, OC 2-6 -alkynyl, C 3 _ 8 -cycloalkyl, C 1-6 -alkyl-C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3- 8-cycloalkyl, aryl, Ci_ 6 -alkylaryl, OC 0-6 -alkylaryl, Ci-e-alkyl-heterocycloalkyl, OCce-alkyl-heterocycloalkyl, (CO)R 10 , O(CO)R 10 , 0(CO)OR
  • B is selected from the group consisting of C 0-6 -alkylaryl and OC 0-6 -alkylaryl, wherein any aryl moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO 2 R, SO 2 R and CN; and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
  • R 1 is phenyl wherein said phenyl is substituted by one or more B;
  • R 2 and R 3 are independently selected from the group consisting of H and C 1-6 -alkyl
  • R 4 is H and R 6 is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, C 1-6 -alkyl, and OCi -6 alkyl;
  • R 5 is selected from the group consisting of H, F, Cl, Br, I, or a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S;
  • R 7 is selected from the group consisting of H or C 1-6 -alkyl
  • R 8 and R 9 are both H
  • B is OCo -6 -alkylaryl, wherein said aryl moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo and alkoxy, and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and n are defined as hereinabove.
  • n is 1, 2, or 3.
  • R 8 and R on adjacent carbon atoms can be missing so as to form partially or fully unsaturated moieties.
  • n is 1 and two adjacent R and R are missing, the moiety is an alkenyl group.
  • R 8 and R 9 are missing, the moiety is an alkynyl group. All of these combinations are contemplated.
  • n is 1.
  • R and R particularly are each H.
  • R is a 5- to 7-membered ring that is selected from the group consisting of aryl, C 3-8- cycloalkyl, cycloalkenyl, and heterocyclyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I 5 OCi -6 -alkylhalo, and OC 0-6 -alkylaryl.
  • Exemplary rings in this context include but are not limited to phenyl, naphthyl, C 3-8- cycloalkyl, cycloalkenyl, furanyl, tetrahydrofuranyl, thiophenyl, pyridyl, oxadiazolyl, quinolinyl, piperazinyl, and tetrahydropyranyl.
  • R 1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I 5 OCi -6 -alkylhalo, and OC 0-6 -alkylaryl.
  • R 1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC 1-6 -alkylhalo, and OC 0-6 -alkylaryl.
  • R 2 , R 3 , R 4 , R 6 , R 8 , and R 9 are each H and n is 1.
  • Particular values for R 7 include H, Cl, Br, I, C 1-6 -alkyl, and OC 1-4 -alkyl, particularly H, Cl, Br, I, -CH 3 , and -OCH 3 , and most particularly Cl 5 Br 5 I, and -OCH 3 .
  • R 1 is a C 3-8 -cycloalkyl group. Particularly, R 1 is cyclopropyl. In this embodiment, n is particularly 1, 2, or 3, and most particularly is 1.
  • R 5 is selected from the group consisting of C 3-8 -cycloalkyl, Ci -6 -alkyl-C 3-8 -cycloalkyl, OC 0-6 -alkyl-C 3-8 -cycloalkyl, aryl, C 1-6 -alkylaryl, OC 1-6 -alkylaryl, and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
  • R 5 may be substituted by one or more A, and any cycloalkyl or aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S.
  • R 5 is selected from Ci_ 6 -alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R may be substituted by one or more A.
  • R 5 is a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, which ring is substituted by one or more A selected from the group consisting of Ci- 6 -alkyl-heterocyclyl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
  • n is 1, 2, or 3;
  • R 4 , R 6 , R 8 and R 9 are each H;
  • R 1 is selected from the group consisting of aryl, C 3-8- cycloalkyl, cycloalkenyl, and heterocyclyl optionally substituted by one or more A selected from the group consisting of F 5 Cl, Br, I, OC t - 6 -alkylhalo, and OC 0-6 -alkylaryl;
  • R 7 is selected from the group consisting of H, Cl, Br, I, C 1-6 -alkyl, and OCi_ 4 -alkyl, and
  • R 5 is selected from the group consisting of C 3- g-cycloalkyl, C 1-6 -alkyl-C 3-8 -cycloalkyl, OCo -6 -alkyl-C 3-8 -cycloalkyl, aryl, C 1-6 -alkylaryl, OCi -6 -alkylaryl, and
  • n is 1, 2, or 3;
  • R 4 , R 6 , R 8 and R 9 are each H;
  • R 1 is selected from phenyl, naphthyl, C 3-8- cycloalkyl, cycloalkenyl, furanyl, tetrahydrofuranyl, thiophenyl, pyridyl, oxadiazolyl, quinolinyl, piperazinyl, and tetrahydropyranyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi -6 -alkylhalo, and OC 0-6 -alkylaryl;
  • R 7 is selected from Cl, Br, I, and -OCH 3
  • R 5 is selected from C 1-6 -alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R 5 may be substituted by one or more
  • n is 1, 2, or 3;
  • R 4 , R 6 , R 8 and R 9 are each H;
  • R 1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC 1-6 -alkylhalo, and OCo -6 -alkylaryl;
  • R 7 is selected from the group consisting of H, Cl, Br, I, C 1-6 -alkyl, and OC 1-4 -alkyl, and
  • R 5 is a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein the 5- to 7-membered ring is substituted by one or more A selected from the group consisting of Ci- 6 -alkyl-heterocyclyl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
  • n is 1 ;
  • R 2 , R 3 , R 4 , R 6 , R 8 and R 9 are each H;
  • R 1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi -6 -alkylhalo, and OC 0-6 -alkylaryl;
  • R 7 is selected from Cl, Br, I, and -OCH 3 , and
  • R 5 is selected from C 1-6 -alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R 5 may be substituted by one or more A.
  • the compounds of the invention may exist in, and be isolated as, enantiomeric or diastereomeric forms, or as a racemic mixture.
  • the present invention includes any possible enantiomers, diastereomers, racemates or mixtures thereof, of a compound of formula I.
  • the optically active forms of the compound of the invention may be prepared, for example, by chiral chromatographic separation of a racemate, by synthesis from optically active starting materials or by asymmetric synthesis based on the procedures described thereafter.
  • salts of the compounds of formula I are also salts of the compounds of formula I.
  • pharmaceutically acceptable salts of compounds of the present invention are obtained using standard procedures well known in the art, for example, by reacting a sufficiently basic compound, for example an alkyl amine with a suitable acid, for example, HCl or acetic acid, to afford a physiologically acceptable anion.
  • a corresponding alkali metal such as sodium, potassium, or lithium
  • an alkaline earth metal such as a calcium
  • a compound of the present invention having a suitably acidic proton, such as a carboxylic acid or a phenol with one equivalent of an alkali metal or alkaline earth metal hydroxide or alkoxide (such as the ethoxide or methoxide), or a suitably basic organic amine (such as choline or meglumine) in an aqueous medium, followed by conventional purification techniques.
  • a suitably acidic proton such as a carboxylic acid or a phenol
  • an alkali metal or alkaline earth metal hydroxide or alkoxide such as the ethoxide or methoxide
  • a suitably basic organic amine such as choline or meglumine
  • the compound of formula I may be converted to a pharmaceutically acceptable salt or solvate thereof, particularly, an acid addition salt such as a hydrochloride, hydrobromide, phosphate, acetate, fumarate, maleate, tartrate, citrate, methanesulphonate or j?-toluenesulphonate.
  • an acid addition salt such as a hydrochloride, hydrobromide, phosphate, acetate, fumarate, maleate, tartrate, citrate, methanesulphonate or j?-toluenesulphonate.
  • compositions include the compounds described herein, their pharmaceutically acceptable salts, hydrates, solvates and optical isomers thereof.
  • the compounds of the present invention may be formulated into conventional pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt or solvate thereof, in associaton with a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • a solid carrier can be one or more substances, which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or table disintegrating agents.
  • a solid carrier can also be an encapsulating material.
  • the carrier is a finely divided solid, which is in a mixture with the finely divided compound of the invention, or the active component.
  • the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • a low-melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient sized moulds and allowed to cool and solidify.
  • Suitable carriers include, but are not limited to, magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, low-melting wax, cocoa butter, and the like.
  • composition is also intended to include the formulation of the active component with encapsulating material as a carrier providing a capsule in which the active component (with or without other carriers) is surrounded by a carrier which is thus in association with it. Similarly, cachets are included.
  • Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration.
  • Liquid form compositions include solutions, suspensions, and emulsions.
  • sterile water or water propylene glycol solutions of the active compounds may be liquid preparations suitable for parenteral administration.
  • Liquid compositions can also be formulated in solution in aqueous polyethylene glycol solution.
  • Aqueous solutions for oral administration can be prepared by dissolving the active component in water and adding suitable colorants, flavoring agents, stabilizers, and thickening agents as desired.
  • Aqueous suspensions for oral use can be made by dispersing the finely divided active component in water together with a viscous material such as natural synthetic gums, resins, methyl cellulose, sodium carboxymethyl cellulose, and other suspending agents known to the pharmaceutical formulation art.
  • Exemplary compositions intended for oral use may contain one or more coloring, sweetening, flavoring and/or preservative agents.
  • the pharmaceutical composition will include from about 0.05%w (percent by weight) to about 99%w, more particularly, from about 0.10%w to 50%w, of the compound of the invention, all percentages by weight being based on the total weight of the composition.
  • a therapeutically effective amount for the practice of the present invention can be determined by one of ordinary skill in the art using known criteria including the age, weight and response of the individual patient, and interpreted within the context of the disease which is being treated or which is being prevented.
  • Neurological and psychiatric disorders amenable to treatment with compounds disclosed herein include, but are not limited to, disorders such as cerebral deficit subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, idiopathic and drug-induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, cerebral deficits secondary to prolonged status epilepticus, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, schizophrenia, anxiety (including generalized anxiety disorder, panic disorder, social phobia, obse
  • the invention thus provides a use of any of the compounds according to formula I, or a pharmaceutically acceptable salt or solvate thereof, for the manufacture of a medicament for the treatment of any of the conditions discussed above.
  • the invention provides a method for the treatment of a subject suffering from any of the conditions discussed above, whereby an effective amount of a compound according to formula I or a pharmaceutically acceptable salt or solvate thereof, is administered to a patient in need of such treatment.
  • the invention also provides a compound of formula I or pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined for use in therapy.
  • the term “therapy” also includes “prophylaxis” unless there are specific indications to the contrary.
  • the term “therapeutic” and “therapeutically” should be construed accordingly.
  • the term “therapy” within the context of the present invention further encompasses the administration of an effective amount of a compound of the present invention, to mitigate either a pre-existing disease state, acute or chronic, or to mitigate a recurring condition.
  • This definition also encompasses prophylactic therapies for prevention of recurring conditions and continued therapy for chronic disorders.
  • the compounds of the present invention may be administered in the form of a conventional pharmaceutical composition by any route including orally, intramuscularly, subcutaneously, topically, intranasally, intraperitoneally, intrathoracially, intravenously, epidurally, intrathecally, intracerebroventricularly and by injection into the joints.
  • the route of administration is oral, intravenous, or intramuscular.
  • the dosage will depend on the route of administration, the severity of the disease, age and weight of the patient and other factors normally considered by the attending physician, who determines the individual regimen and dosage level for a particular patient.
  • the compounds described herein may be provided or delivered in a form suitable for oral use, for example, in a tablet, lozenge, hard and soft capsule, aqueous solution, oily solution, emulsion, and suspension.
  • the compounds may be formulated into a topical administration, for example, as a cream, ointment, gel, spray, or aqueous solution, oily solution, emulsion or suspension.
  • the compounds described herein also may be provided in a form that is suitable for nasal administration, for example, as a nasal spray, nasal drops, or dry powder.
  • the compounds can be administered to the vagina or rectum in the form of a suppository.
  • the compounds described herein also may be administered parentally, for example, by intravenous, intravesicular, subcutaneous, or intramuscular injection or infusion.
  • the compounds can be administered by insufflation (for example as a finely divided powder).
  • the compounds may also be administered transdermally or sublingually.
  • the compounds of formula I, or salts thereof are useful as pharmacological tools in the development and standardisation of in vitro and in vivo test systems for the evaluation of the effects of inhibitors of mGluR-related activity in laboratory animals as part of the search for new therapeutics agents.
  • Such animals include, for example, cats, dogs, rabbits, monkeys, rats and mice. Processes for Preparing:
  • a compound of formula Ib is then cross-coupled with a suitable reagent containing R to yield a a compound according to formula Ic:
  • 5-substituted-7-methyl isoindolones are synthesized as depicted in Scheme 1 below.
  • 4-bromo-2,6-dimethylaniline is converted to the corresponding nitrile under Sandmeyer reaction conditions.
  • the nitrile is then hydrolyzed to the acid in a stepwise fashion.
  • the amide can be obtained by basic hydrolysis.
  • the amide is then diazotized and hydrolyzed with nitrososulphuric acid to provide the benzoic acid, which is subsequently protected as the methyl ester using standard conditions.
  • the benzylic methyl group is monobrominated with N-bromosuccinimide using benzoyl peroxide as the radical initiator.
  • 5-substituted-7-chloro isoindolones are synthesized as depicted in Scheme 2 below.
  • 4-bromo-2-methylbenzoic acid is chlorinated ortho to the acid using N-chlorosuccinimide and a palladium catalyst.
  • this acid was then esterified, brominated, and cyclized to yield the isoindolone intermediate.
  • Substituent R 5 is introduced similarly.
  • isoindolones that are substituted with an amide at C5 can be prepared as depicted in Scheme 3 below.
  • an appropriately substituted 5-bromoisoindolone is converted to the corresponding nitrile using zinc cyanide in the presence of a palladium catalyst.
  • the nitrile is then hydrolyzed under basic conditions to provide the benzoic acid, which was then coupled with various amines using methodologies that are well-known in the art to provide the final compounds.
  • the pharmacological properties of the compounds of the invention can be analyzed using standard assays for functional activity.
  • glutamate receptor assays are well known in the art as described in, for example, Aramori et ah, 1992, Neuron, 8:757; Tanabe et al, 1992, Neuron, 8:169; Miller etal, 1995, J. Neuroscience, 15:6103; Balazs, et al, 1997, J. Neurochemistry, 1997,69:151.
  • the methodology described in these publications is incorporated herein by reference.
  • the compounds of the invention can be studied by means of an assay that measures the mobilization of intracellular calcium, [Ca 2+ ] j in cells expressing mGluR2.
  • Fluorometric Imaging Plate Reader FLIPR analysis was used to detect allosteric activators of mGluR2 via calcium mobilization.
  • FLIPR Fluorometric Imaging Plate Reader
  • the cells were trypsinized and plated in DMEM at 100,000 cells/well in black sided, clear-bottom, collagen I coated, 96-well plates. The plates were incubated under 5% CO 2 at 37 0 C overnight. Cells were loaded with 6 ⁇ M fluo-3 acetoxymethylester (Molecular Probes, Eugene Oregon) for 60 minutes at room temperature.
  • FLIPR experiments were done using a laser setting of 0.8 W and a 0.4 second CCD camera shutter speed. Extracellular fluo-3 was washed off and cells were maintained in 160 ⁇ L of buffer and placed in the FLIPR. An addition of test compound (0.01 ⁇ M to 30 ⁇ M in duplicate) was made after 10 seconds of baseline fluorescent readings were recorded on FLIPR. Fluorescent signals were then recorded for an additional 75 seconds at which point a second addition of DCG-IV (0.2 ⁇ M) was made and fluorescent signals were recorded for an additional 65 seconds. Fluorescent signals were measured as the peak height of the response within the sample period. Data was analyzed using Assay Explorer, and EC 50 and E max values (relative to maximum DCG-IV effect) were calculated using a four parameter logistic equation.
  • a [ 35 S]-GTPyS binding assay was used to functionally assay mGluR2 receptor activation.
  • the allosteric activator activity of compounds at the human mGluR2 receptor were measured using a [ 35 S]-GTPyS binding assay with membranes prepared from CHO cells which stably express the human mGluR2.
  • the assay is based upon the principle that agonists bind to G-protein coupled receptors to stimulate GDP-GTP exchange at the G-protein. Since [ 35 S]-GTPyS is a non-hydrolyzable GTP analog, it can be used to provide an index of GDP-GTP exchange and, thus, receptor activation.
  • the GTPyS binding assay therefore provides a quantitative measure of receptor activation.
  • Membranes were prepared from CHO cells stably transfected with human mGluR2. Membranes (30 ⁇ g protein) were incubated with test compound (3 nM to 300 ⁇ M) for 15 minutes at room temperature prior to the addition of 1 ⁇ M glutamate, and incubated for 30 min at 30 0 C in 500 ⁇ L assay buffer (20 mM HEPES, 100 mM NaCl, 10 raM MgCl 2 ), containing 30 ⁇ M GDP and 0.1 nM [ 35 S]-GTPyS (1250 Ci/mmol). Reactions were carried out in triplicate in 2 mL polypropylene 96-well plates.
  • Example 1 5-Bromo-2-[4-(2-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Psychology (AREA)
  • Hospice & Palliative Care (AREA)
  • Vascular Medicine (AREA)
  • Anesthesiology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Otolaryngology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Indole Compounds (AREA)

Abstract

Compounds of formula (I), wherein R1 is a ring , and uses of the compounds in therapy as metabotropic glutamate receptors modulators, particularly in neurological and psychiatric disorders.

Description

METABOTROPIC GLUTAMATE-RECEPTOR-POTENTIATING ISOINDOLONES
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the priority of P.C.T. Application PCT/US2005/028760, filed August 12, 2005.
BACKGROUND OF THE INVENTION
The present invention relates to novel compounds that function as potentiators of glutamate receptors, methods for their preparation, pharmaceutical compositions containing them and their use in therapy.
The metabotropic glutamate receptors (mGluR) are a family of GTP -binding-protein (G-protein) coupled receptors that are activated by glutamate, and that have important roles in synaptic activity in the central nervous system, neural plasticity, neural development and neurodegeneration.
Activation of mGluRs in intact mammalian neurons elicits one or more of the following responses: activation of phospholipase C; increases in phosphoinositide (PI) hydrolysis; intracellular calcium release; activation of phospholipase D; activation or inhibition of adenyl cyclase; increases or decreases in the formation of cyclic adenosine monophosphate (cAMP); activation of guanylyl cyclase; increases in the formation of cyclic guanosine monophosphate (cGMP); activation of phospholipase A2; increases in arachidonic acid release; and increases or decreases in the activity of voltage- and ligand-gated ion channels (Schoepp et al, 1993, Trends Pharmacol. Sci., 14:13 ; Schoepp, 1994, Neurochem. Int., 24:439; Pin et al, 1995, Neuropharmacology 34:1; Bordi & Ugolini, 1999, Prog. Neurobiol. 59:55).
Eight mGluR subtypes have been identified. The subtypes are divided into three groups based upon primary sequence similarity, signal transduction linkages, and pharmacological profile. Group-I includes mGluRl and mGluR5, which activate phospholipase C and the generation of an intracellular calcium signal. Group-II (mGluR2 and niGluR3) and Group-Ill (mGluR4, rnGluRό, mGluR7, and mGluR8) mGluRs mediate an inhibition of adenylyl cyclase activity and cyclic AMP levels. For a review, see Pin et ah, 1999, Eur. J. Pharmacol., 375:277-294. Activity of mGluR family receptors is implicated in a number of normal processes in the mammalian CNS, and are important targets for compounds for the treatment of a variety of neurological and psychiatric disorders. Activation of mGluRs is required for induction of hippocampal long-term potentiation and cerebellar long-term depression (Bashir et ah, 1993, Nature, 363:347 ; Bortolotto et a!., 1994, Nature, 368:740 ; Aiba et al., 1994, Cell, 79:365 ; Aiba et ah, 1994, Cell, 79:377). A role for mGluR activation in nociception and analgesia also has been demonstrated (Meller et ah, 1993, Neuroreport, 4: 879; Bordi & Ugolini, 1999, Brain Res., 871 :223). In addition, mGluR activation has been suggested to play a modulatory role in a variety of other normal processes including synaptic transmission, neuronal development, apoptotic neuronal death, synaptic plasticity, spatial learning, olfactory memory, central control of cardiac activity, waking, motor control and control of the vestibulo-ocular reflex (Nakanishi, 1994, Neuron, 13:1031; Pin et ah, 1995, Neuropharmacology, see above; Knopfel et al., 1995, J. Med. Chem., 38:1417).
Recent advances in the elucidation of the neurophysiological roles of mGluRs have established these receptors as promising drug targets in the therapy of acute and chronic neurological and psychiatric disorders and chronic and acute pain disorders. Because of the physiological and pathophysiological significance of the mGluRs, there is a need for new drugs and compounds that can modulate mGluR function.
SUMMARY OF THE INVENTION
We have identified a class of compounds that modulate mGluR function. In one aspect the invention provides compounds of formula I,
Figure imgf000003_0001
wherein:
R1 is a 3- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said ring is substituted by one or more B; R2 and R3 are independently selected from the group consisting of H, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, heterocycloalkyl, Cs.g-cycloalkyl, Ci-6-alkyl-aryl, Ci-6-alkyl-heteroaryl,
Figure imgf000004_0001
and Ci-β-alkyl-Cs-s-cycloalkyl, wherein R and R3 may be substituted by one or more A;
R4 and R6 are independently selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, Ci-6-alkyl, Q-e-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C^s-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8~cycloalkyl, aryl, C1-6-alkylaryl, OC0.6-alkylaryl, (CO)R10, Q(CO)R10, 0(CO)OR10, C(O)OR10, 0(CNR10)ORπ, C1-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, C0-6-alkylCO2R10, OC,.6-alkylCO2R10, d-e-alkylcyano, OC2-6-alkylcyano, C0-6-alkylNR10Rπ, OC2-6-alkylNR10Rπ, C1.6-alkyl(CO)NR10R11, OC1-6-alkyl(CO)NR10R11, C0-6-alkylNR10(CO)Rn, OC2-6-alkylNR10(CO)R11, Co-6-alkylNR10(CO)NR10Rπ, C0.6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, Co-e-alkyKSO^NR'V ' , OC2-6-alkyl(SO2)NR10R1 ' , C0-6-alkylNR10(SO2)R11 , OC2-6-alkylNR10(SO2)Rn, Co.6-alkylNR10(S02)NR10Rn, OC2-6-alkylNR10(SO2)NR10R1 ', (CO)NR10R11 , 0(CO)NR10R1 ' , NR10OR1 * , Co-6-alkylNR1 °(CO)OR' ' , OC2-6-alkylNR10(CO)ORn, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R4 and R6 may be substituted by one or more A, and wherein any cycloalkyl or aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci_6-alkyl, OCo-6-alkyl, Ci_6-alkylhalo, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3_8-cycloalkyl, Ci-e-alkyl-Cs-s-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, C1-6-alkylheteroaryl, OCi-6-alkylaryl, OC1-6-alkylheteroaryl, Ci-ό-alkylheterocycloalkyl, Oheterocycloalkyl, OQ-e-alkylheterocycloalkyl, C(O)H, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, 0(CN)OR10, C^-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, Co-6-alkylC02R10, Cμe-alkylcyano, OC2-6-alkylcyano, C0-6-alkylNR10RH, OC2-6-alkylNR10Rn, Ci-6-alkyl(CO)NR10Rπ, OC1-6-alkyl(CO)NR10R11, Co-6-alkylNR10(CO)R11, OC2-6-alkylNRIO(CO)R11, Co-6-alkylNR10(CO)NRIOR", C0-6-alkylSR10, OC2-6-alkylSR10, C0.6-alkyl(SO)R10, OC2-6-BIlCyI(SO)R1 °, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, C0-6-alkyl(SO2)NRI0Ru, OC2-6-alkyl(SO2)NR10R1 ', Qw-alkylNR10CSO2)R1 ', OC^-alkylNR'^SO^R1 ',
Figure imgf000005_0001
OC2-6-alkylNRI0(SO2)NR10Rπ, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNR10(CO)OR11, OC2-6-alkylNR10(CO)ORπ, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi-4-alkyl, Ci-6-alkyl, C1-6-alkylhalo, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, and C3-8-cycloalkyl;
R8 and R9 are independently selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, C1-6-alkylhalo, OC1-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl,
OC2-6-alkenyl, C2-6-alkynyl, and OC2-6-alkynyl, or, where n is greater than 1 , two or more R and/or R9 on adjacent carbon atoms may be absent to form an alkenyl or alkynyl moiety;
R1 and R l are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, Ci-6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OC0-6-alkylaryl, Co-β-alkyl-heterocycloalkyl, OCj-β-alkyl-heterocycloalkyl, heteroaryl, and Ci-6alkylheteroaryl, wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S and any cyclic moiety is optionally substituted with a substituent selected from alkyl, halo, hydroxyl, Oalkyl, haloalkyl and Ohaloalkyl;
A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, Ci-6-alkyl, Ci-6-alkylhalo, OC1-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OCo-β-alkyl-Cs-s-cycloalkyl, aryl, Ci-6-alkylaryl, OCo-6-alkylaryl, d-δ-alkyl-heterocyclyl, Ci-e-alkyl-heterocycloalkyl, OC0-6-alkyl-heterocycloalkyl, (CO)R10, 0(CO)R10, 0(CO)OR10, O(CNR10)ORπ, C1-6-alkylOR10, OC2-6-alkylOR10, Ci-6-alkyl(CO)R10, OC,-6-alkyl(CO)R10, C0-6-alkylCO2R10, Od-e-alkylCOzR10, Ci.6-alkylcyano, OC2-6-alkylcyano, C0-6-alkylNR10Ru,
Figure imgf000006_0001
C0-6-alkyl(CO)NRI0R11,
Figure imgf000006_0002
Co-e-alkylNR10CCO)R11 , OC2-6-alkylNR10CCO)R1 ] , Co-e-alkylNR1 °(CO)NR' 0R1 ' , C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2.6-alkyl(SO)R10, Ci-6-alkylSO2R10, OC2-6-alkylSO2R10, C0-6-alkyl(SO2)NR10R1 ' , OC2-6-alkyl(SO2)NR' 0R1 ' , Co-β-alkylNR10CSO2)R1 ', OC2-6-alkylNR10(SO2)R1 ',
Figure imgf000006_0003
OC^e-alkylNR'^SO^NR^R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNR10(CO)OR11, OC2-6-alkylNR10(CO)ORn, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said 5- to 7-membered ring is optionally substituted by one or more of R10 and R11;
B is selected from the group consisting of C3-8-cycloalkyl, C1-6-alkyl-C3-g-cycloalkyl, OCo-β-alkyl-CB-s-cycloalkyl, C0-6-alkylaryl, OC0-6-alkylaryl, Ci-β-alkyl-heterocycloalkyl, C1-6-alkyl-heterocycloalkyl, OCo-β-alkyl-heterocycloalkyl, C0-6-alkyl-heteroaryl and OC0-6-alkyl-heteroaryl, wherein any cyclic moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO2R, SO2R and CN; and n is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof, with a proviso that said compound is not: 2- [4-(4-fluoro-phenoxy)-benzyl] -2,3 -dihydro-isoindol- 1 -one; 2-[4-(4-trifluoromethyl-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one; 2-{3-[4-(4-fluoro-phenoxy)-phenyl]-propyl}-7-iodo-2,3-dihydro-isoindol-l-one, or 7-iodo-2-[3-(2-methoxy-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one.
The invention also provides processes for the preparation of compounds of formula I.
The invention further provides a pharmaceutical composition comprising a compound according to formula I together with a pharmaceutically acceptable carrier or excipient; in another aspect the invention provides a method for the treatment or prevention of neurological and psychiatric disorders associated with glutamate dysfunction in an animal in need of such treatment. The method comprises the step of administering to the animal a therapeutically effective amount of a compound of formula I or a pharmaceutical composition thereof.
The invention also provides for the use of a compound according to formula I, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for the treatment of any of the conditions discussed herein. Further, the invention provides a compound of formula I, or a pharmaceutically acceptable salt or solvate thereof, for use in therapy.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is based upon the discovery of compounds that exhibit activity as modulators of metabotropic glutamate receptors. More particularly, the compounds of the present invention exhibit activity as potentiators of the mGluR2 receptor, and are useful in therapy, in particular as pharmaceuticals for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction.
The disclosure of P.C.T. Application PCT/US2005/028760 is incorporated herein by reference in its entirety. Definitions
Unless specified otherwise within this specification, the nomenclature used in this specification generally follows the examples and rules stated in Nomenclature of Organic Chemistry, Sections A, B, C, D, E, F, and H, Pergamon Press, Oxford, 1979, which is incorporated by references herein for its exemplary chemical structure names and rules on naming chemical structures. Optionally, a name of a compound may be generated using a chemical naming program: ACD/ChemSketch, Version 5.09/September 2001, Advanced Chemistry Development, Inc., Toronto, Canada.
The term "alkyl" as used herein means a straight- or branched-chain hydrocarbon radical having, for example, from one to six carbon atoms, and includes methyl, ethyl, propyl, isopropyl, t-butyl and the like.
The term "alkenyl" as used herein means a straight- or branched-chain alkenyl radical having, for example, from two to six carbon atoms, and includes ethenyl, 1 -propenyl, 1-butenyl and the like. The term "alkynyl" as used herein means a straight- or branched-chain alkynyl radical having, for example, from two to six carbon atoms, and includes 1-propynyl (propargyl), 1-butynyl and the like.
The term "cycloalkyl" as used herein means a cyclic group (which may be unsaturated) having, for example, from three to seven carbon atoms, and includes cyclopropyl, cyclohexyl, cyclohexenyl and the like.
The term "heterocycloalkyl" as used herein means, for example, a three- to seven-membered cyclic group (which may be unsaturated) having at least one heteroatom selected from the group consisting of N, S and O, and includes piperidinyl, piperazinyl, pyrrolidinyl, tetrahydrofuranyl and the like.
The term "alkoxy" as used herein means a straight- or branched-chain alkoxy radical having, for example, from one to six carbon atoms and includes methoxy, ethoxy, propyloxy, isopropyloxy, t-butoxy and the like.
The term "halo" as used herein means halogen and includes fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms.
The term "aryl" as used herein means an aromatic group having, for example, five to twelve atoms, and includes phenyl, naphthyl and the like.
The term "heteroaryl" means an aromatic group which includes at least one heteroatom selected from the group consisting of N, S and O, and includes groups and includes pyridyl, indolyl, furyl, benzofuryl, thienyl, benzothienyl, quinolyl, oxazolyl and the like.
The term "alkanoyl" as used herein means a straight- or branched-chain alkanoyl radical having, for example, from two to seven atoms, and includes acetyl, propionyl, butyryl and the like.
The term "cycloalkenyl" as used herein means an unsaturated cylcloaklyl group having, for example, from four to seven carbon atoms, and includes cyclopent-1-enyl, cyclohex-1-enyl and the like.
The terms "alkylaryl", "alkylheteroaryl " and "alkylcycloalkyl " refer to an alkyl radical substituted with an aryl, heteroaryl or cycloalkyl group, and includes 2-phenethyl, 3 -cyclohexyl propyl and the like. The term "5- to 7-membered ring that may contain one or more heteroatoms independently selected from N, O and S" includes aromatic and heteroaromatic rings, as well as carbocyclic and heterocyclic rings which may be saturated or unsaturated, and includes furyl, isoxazolyl, oxazolyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, thiazolyl, thienyl, imidazolyl, triazolyl, morpholinyl, piperazinyl, piperidinyl, homopiperidinyl, tetrahydropyranyl, phenyl, cyclohexyl, cycloheptyl, cyclopentyl, cyclohexanyl and the like.
The term "pharmaceutically acceptable salt" means either an acid addition salt or a basic addition salt which is compatible with the treatment of patients.
A "pharmaceutically acceptable acid addition salt" is any non-toxic organic or inorganic acid addition salt of the base compounds represented by Formula I or any of its intermediates. Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids which form suitable salts include the mono-, di- and tricarboxylic acids. Illustrative of such acids are, for example, acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, salicylic, 2-phenoxybenzoic, p-toluenesulfonic acid and other sulfonic acids such as methanesulfonic acid and 2-hydroxyethanesulfonic acid. Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated, solvated or substantially anhydrous form. In general, the acid addition salts of these compounds are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms. The selection criteria for the appropriate salt will be known to one skilled in the art. Other non-pharmaceutically acceptable salts e.g. oxalates may be used for example in the isolation of compounds of Formula I for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
A "pharmaceutically acceptable basic addition salt" is any non-toxic organic or inorganic base addition salt of the acid compounds represented by Formula I or any of its intermediates. Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxides. Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethyl amine and picoline or ammonia. The selection of the appropriate salt may be important so that an ester functionality, if any, elsewhere in the molecule is not hydrolyzed. The selection criteria for the appropriate salt will be known to one skilled in the art.
"Solvate" means a compound of Formula I or the pharmaceutically acceptable salt of a compound of Formula I wherein molecules of a suitable solvent are incorporated in a crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered as the solvate. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a hydrate.
The term "stereoisomers" is a general term for all isomers of the individual molecules that differ only in the orientation of their atoms in space. It includes mirror image isomers (enantiomers), geometric (cis/trans) isomers and isomers of compounds with more than one chiral centre that are not mirror images of one another (diastereomers).
The term "treat" or "treating" means to alleviate symptoms, eliminate the causation of the symptoms either on a temporary or permanent basis, or to prevent or slow the appearance of symptoms of the named disorder or condition.
The term "therapeutically effective amount" means an amount of the compound which is effective in treating the named disorder or condition.
The term "pharmaceutically acceptable carrier" means a non-toxic solvent, dispersant, excipient, adjuvant or other material which is mixed with the active ingredient in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to the patient. One example of such a carrier is a pharmaceutically acceptable oil typically used for parenteral administration. Compounds:
Compounds of the invention conform generally to formula I:
Figure imgf000010_0001
wherein:
R1 is a 3- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said ring is substituted by one or more B; R2 and R3 are independently selected from the group consisting of H, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, heterocycloalkyl, C3-8-cycloalkyl, Ci-6-alkyl-aryl, Ci_6-alkyl-heteroaryl, Ci_6-alkyl-heterocycloalkyl, and Ci-6-alkyl-C3-8-cycloalkyl, wherein R2 and R3 may be substituted by one or more A;
R4 and R6 are independently selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, Ci-6-alkylhalo, OC1-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-Cy cloalkyl, Ci-δ-alkyl-Cs.s-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OC0-6-alkylaryl, (CO)R10, O(CO)R10, 0(CO)OR10, C(O)OR10, O(CNR10)ORn, C1-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OCi-6-alkyl(CO)R10, Co-6-alkylC02R10, OCi-6-alkylCO2R10, d-6-alkylcyano, OC2-6-alkylcyano, Co-e-alkylNR^R11, OC2-6-alkylNR10Ru, C1-6-^yI(CO)NR10R1 ', OCi-6-^yI(CO)NR10R1 \ Ccg-alkylNR1 ^CO)R1 \ OC2.6-alkylNR10(CO)R11, C0.6-alkylNR10(CO)NR10R11, C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, C0-6-alkyl(SO2)NR10R11 , OC2-6-alkyl(SO2)NR10R11 , Co-e-alkylNR1 °(SO2)R' l , OC2-6-alkylNR10(S02)R11, Co-6-alkylNR10(S02)NR10R11, OC2-6-alkylNR10(S02)NR10R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, Co^-alkylNR1 °(CO)OR1 ', OC2-6-alkylNR10(CO)OR11, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R4 and R6 may be substituted by one or more A, and wherein any cycloalkyl or aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci-6-alkyl, OCo-6-alkyl, C1-6-alkylhalo, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3.g-cycloalkyl, Ci-β-alkyl-Cs-s-cycloalkyl, OCo-o-alkyl-Cs-s-cycloalkyl, aryl, Cj-6-alkylaryl, C1-6-alkylheteroaryl, OC1-6-alkylaryl, OC1-6-alkylheteroaryl, Ci-β-alkylheterocycloalkyl, Oheterocycloalkyl, OC^-alkylheterocycloalkyl, C(O)H, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, 0(CN)OR10, Ci-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, C0-6-alkylCO2R10, C1-6-alkylcyano, OC2-6-alkylcyano, C^-alkylNR^R11, OC2-6-alkylNR10Rn, C1-6-alkyl(CO)NR10Rπ, OC1-6-alkyl(CO)NR10R11, C0-6-alkylNR10(CO)R11, OC2-6-alkylNR10(CO)R11, C0.6-alkylNR10(CO)NR10Ru, C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, C0-6-alkyl(SO2)NRI0Rπ, OC2-6-alkyl(SO2)NR10Rn, C^-alkylNR1 ^SO2)R1 ', OC2-6-alkylNR10(SO2)Rπ, Co.e-alkylNR^SO^NR'V1, OQj.e-alkylNR1 ^SO2)NR10R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, Co-e-alkylNR10CCO)OR1 ', OC2-6-alkylNR10(CO)ORπ, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi-4-alkyl, Ci-6-alkyl, C1-6-alkylhalo, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, and C3-s-cycloalkyl; o Q
R and R are independently selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, C1-6-alkylhalo, OC1-6alkyl, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, and OC2-6-alkynyl, or, where n is greater than 1 , two or more R and/or R9 on adjacent carbon atoms may be absent to form an alkenyl or alkynyl moiety;
R10 and R11 are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, Ci-6-alkylhalo, OC1-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci-6-alkyl-C3-g-cycloalkyl, OCo-6-alkyl-Cs-s-cycloalkyl, aryl, Ci-6-alkylaryl, OC0-6-alkylaryl, Co-e-alkyl-heterocycloalkyl, OCi-β-alkyl-heterocycloalkyl, heteroaryl, and Ci-6alkylheteroaryl, wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S and any cyclic moiety is optionally substituted with a substituent selected from alkyl, halo, hydroxyl, Oalkyl, haloalkyl and Ohaloalkyl;
A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, C1-6-alkyl, C1-6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OC0-6-alkylaryl, Cj-e-alkyl-heterocyclyl, Ci-e-alkyl-heterocycloalkyl, OCo-e-alkyl-heterocycloalkyl, (CO)R10, 0(CO)R10, 0(CO)OR10, O(CNR10)ORn, Ci-6-alkylOR10, OC2-6-alkylOR10, Ci-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, C0-6-alkylCO2R10, OCi-6-alkylCO2R10, CI-6-alkylcyano, OC2-6-alkylcyano, C0.6-alkylNR10Rπ 3 OC2-6-^yINR10R1 \ C0-6-alkyl(CO)NR10R1 ', OCi-6-alkyl(CO)NR10R1 \ C0-6-alkylNR10(CO)R1 ', OC2-6-alkylNR10(CO)R1 ', C0-6-alkylNR10(CO)NR10R1 ', C0.6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C1-6-alkylSO2R10, OC2-6-alkylSO2R10, C0-6-^yI(SO2)NR10R11, OC2-6-alkyl(SO2)NR10Rπ 5
Figure imgf000013_0001
OCz.e-alkylNR'^SO^NR^R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNR10(CO)OR11, OQ^-alkylNR1 ^CO)OR1 \ SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said 5- to 7-membered ring is optionally substituted by one or more of R10 and R11;
B is selected from the group consisting of C^s-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, C0-6-alkylaryl, OC0-6-alkylaryl,
Figure imgf000013_0002
Ci-e-alkyl-heterocycloalkyl, OCo-β-alkyl-heterocycloalkyl, C0-6-alkyl-heteroaryl and OCo-6-alkyl-heteroaryl, wherein any cyclic moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO2R, SO2R and CN, and n is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof, with a proviso that said compound is not: 2-[4-(4-fluoro-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one; 2-[4-(4-trifluoromethyl-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one; 2-{3-[4-(4-fluoro-phenoxy)-phenyl]-propyl}-7-iodo-2,3-dihydro-isoindol-l-one, or 7-iodo-2-[3-(2-methoxy-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one.
Particular compounds according to Formula I, are those wherein:
R1 is phenyl wherein said phenyl is substituted by one or more B;
R2 and R3 are independently selected from the group consisting of H, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, heterocycloalkyl, C3-8-cycloalkyl, Ci-6-alkyl-aryl, C1-6-alkyl-heteroaryl, Ci-e-alkyl-heterocycloalkyl, and C1-6-alkyl-C3-8-cycloalkyl, wherein R2 and R3 may be substituted by one or more A; R4 and R6 are independently selected from the group consisting of H5 hydroxy, F, Cl, Br, I, nitro, cyano, Ci-6-alkyl, Ci-6-alkylhalo, OCi.6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci.6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, OC0-6-alkylaryl, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, O(CNR10)ORn, Ci-6-alkylOR10, OC2-6-alkylOR10, Ci-6-alkyl(CO)R10, OCi-6-alkyl(CO)R10, C0-6-alkylCO2R10, OCi-6-alkylCO2R10, Ci.6-alkylcyano, OC2-6-alkylcyano, Co-6-alkylNRloRn, OC2-6-alkylNR10Rn, Ci-6-alkyl(CO)NR10R11, OC1-6-alkyl(CO)NR10R11, C0-6-alkylNR10(CO)R11, OC2-6-alkylNR10(CO)R1 1, C0-6-alkylNR10(CO)NRI0R1 1, C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2.6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10 5 Co-6-alkyl(S02)NR10R11, OC2-6-alkyl(S02)NR10R11, Co-6-alkylNR10(S02)R1 1, OC2-6-alkylNR10(S02)R11, Co-6-alkylNR10(S02)NR10R1 I, OC2-6-alkylNRIO(S02)NR10R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, Co-β-alkylNR1 ^CO)OR1 ', OC2-6-alkylNR10(CO)OR11 ) SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R4 and R6 may be substituted by one or more A, and wherein any cycloalkyl or aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci-6-alkyl, OCo-6-alkyl, Ci-6-alkylhalo, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl,
Figure imgf000014_0001
OC0-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, Ci-6-alkylheteroaryl, OC1-6-alkylaryl, OC1-6-alkylheteroaryl, Ci-β-alkylheterocycloalkyl, Oheterocycloalkyl, OCi-e-alkylheterocycloalkyl, C(O)H, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, 0(CN)OR10, Ci-6-alkylOR10, OC2-6-alkylOR10, Ci-6-alkyl(CO)R10, OC]-6-alkyl(CO)R10, C0-6-alkylCO2R10, Ci-6-alkylcyano, OC2-6-alkylcyano, C0-6-alkylNR10Rn, OC2-6-alkylNR10Rπ, C1-6-alkyl(CO)NR10Ru 5 OCi-6-alkyl(CO)NR10Rn, C0-6-alkylNR10(CO)R11, OC2-6-alkylNR10(CO)R1 1, C0-6-alkylNR10(CO)NR10R11, C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, Co-6-alkyl(S02)NR10Rπ, OC2-6-alkyl(SO2)NR10R1 \ C0-6-alkylNR10(SO2)R11 , OC2-6-alkylNR10(SO2)R1 ' , C0-6-alkylNR10(SO2)NR10R11, OC2-6-alkylNR10(SO2)NR10Rn, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNR10(CO)ORu, OQ^-alkylNR1 ^CO)OR1 ', SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi-4-alkyl, Ci-6-alkyl, Ci-6-alkylhalo, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, and C3-8-cycloalkyl;
R8 and R9 are both H;
R10 and R11 are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, Ci-6-alkyl, Ci_6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci.6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OC0-6-alkylaryl, Co-ό-alkyl-heterocycloalkyl, OCi-o-alkyl-heterocycloalkyl, heteroaryl, and C1-6alkylheteroaryl, wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S and any cyclic moiety is optionally substituted with a substituent selected from alkyl, halo, hydroxyl, Oalkyl, haloalkyl and Ohaloalkyl;
A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, Ci-6-alkyl, C1-6-alkylhalo, OC1-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3_8-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, Ci_6-alkylaryl, OC0-6-alkylaryl,
Figure imgf000015_0001
Ci-e-alkyl-heterocycloalkyl, OCce-alkyl-heterocycloalkyl, (CO)R10, O(CO)R10, 0(CO)OR10, 0(CNR10PR1 ', C1-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OCi-6-alkyl(CO)R10, C0-6-alkylCO2R10, OC1-6-alkylCO2R10, Ci-6-alkylcyano, OC2-6-alkylcyano, Cce-alkylNR^R11, OC2-6-^yINR10R1 ', C0-6-^yI(CO)NR10R1 \ OC1-6-^yI(CO)NR10R1 \ C0-6-alkylNR10(CO)R1 \ OC2-6-alkylNRI0(CO)R1 ', C0-6-alkylNR10(CO)NR10R1 \ Co-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, Ci-6-alkylSO2R10, OC2-6-alkylS02R10, Co-6-alkyl(S02)NR10R11, OC2-6-alkyl(SO2)NR10Rn, Co-6-alkylNR10(S02)R1 \ OC2-6-alkylNR10(SO2)R1 !, C0-6-alkylNR10(SO2)NR10R1 ] , OC2-6-alkylNR10(SO2)NR10Rπ, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNR10(CO)OR11, OC2-6-alkylNR10(CO)ORn, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said 5- to 7-membered ring is optionally substituted by one or more of R10 and R11;
B is selected from the group consisting of C0-6-alkylaryl and OC0-6-alkylaryl, wherein any aryl moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO2R, SO2R and CN; and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
Other particular compounds according to Formula I, are those wherein:
R1 is phenyl wherein said phenyl is substituted by one or more B;
R2 and R3 are independently selected from the group consisting of H and C1-6-alkyl;
R4 is H and R6 is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, and OCi-6alkyl;
R5 is selected from the group consisting of H, F, Cl, Br, I, or a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S;
R7 is selected from the group consisting of H or C1-6-alkyl;
R8 and R9 are both H;
B is OCo-6-alkylaryl, wherein said aryl moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo and alkoxy, and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
Other compounds of the invention conform generally to formula I: wherein R1, R2, R3, R4, R5, R6, R7, R8, R9 and n are defined as hereinabove. In a particular embodiment, n is 1, 2, or 3. When n is greater than 1, two or more of R8 and R on adjacent carbon atoms can be missing so as to form partially or fully unsaturated moieties. Thus, for example, when n is 2 and two adjacent R and R are missing, the moiety is an alkenyl group. When four adjacent R8 and R9 are missing, the moiety is an alkynyl group. All of these combinations are contemplated. Most particularly, n is 1. In this context, R and R particularly are each H.
Another particular subset of compounds are those in which R and R in formula I are each H. Thus, the aromatic portion of the isoindolone core in this embodiment can be di-substituted at most.
In another embodiment, R is a 5- to 7-membered ring that is selected from the group consisting of aryl, C3-8-cycloalkyl, cycloalkenyl, and heterocyclyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I5 OCi-6-alkylhalo, and OC0-6-alkylaryl. Exemplary rings in this context include but are not limited to phenyl, naphthyl, C3-8-cycloalkyl, cycloalkenyl, furanyl, tetrahydrofuranyl, thiophenyl, pyridyl, oxadiazolyl, quinolinyl, piperazinyl, and tetrahydropyranyl. Particularly, R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I5 OCi-6-alkylhalo, and OC0-6-alkylaryl.
In another embodiment, R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC1-6-alkylhalo, and OC0-6-alkylaryl. Additionally, R2, R3, R4, R6, R8, and R9 are each H and n is 1. Particular values for R7 include H, Cl, Br, I, C1-6-alkyl, and OC1-4-alkyl, particularly H, Cl, Br, I, -CH3, and -OCH3, and most particularly Cl5 Br5 I, and -OCH3.
In yet another embodiment, R1 is a C3-8-cycloalkyl group. Particularly, R1 is cyclopropyl. In this embodiment, n is particularly 1, 2, or 3, and most particularly is 1.
Another particular subset of compounds are those in which R5 is selected from the group consisting of C3-8-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OC1-6-alkylaryl, and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S. In this embodiment, R5 may be substituted by one or more A, and any cycloalkyl or aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S. Particularly, R5 is selected from Ci_6-alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R may be substituted by one or more A. More particularly, R5 is a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, which ring is substituted by one or more A selected from the group consisting of Ci-6-alkyl-heterocyclyl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
In yet another embodiment, n is 1, 2, or 3; R4, R6, R8 and R9 are each H; R1 is selected from the group consisting of aryl, C3-8-cycloalkyl, cycloalkenyl, and heterocyclyl optionally substituted by one or more A selected from the group consisting of F5 Cl, Br, I, OCt-6-alkylhalo, and OC0-6-alkylaryl; R7 is selected from the group consisting of H, Cl, Br, I, C1-6-alkyl, and OCi_4-alkyl, and R5 is selected from the group consisting of C3-g-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OCi-6-alkylaryl, and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S.
In another embodiment, n is 1, 2, or 3; R4, R6, R8 and R9 are each H; R1 is selected from phenyl, naphthyl, C3-8-cycloalkyl, cycloalkenyl, furanyl, tetrahydrofuranyl, thiophenyl, pyridyl, oxadiazolyl, quinolinyl, piperazinyl, and tetrahydropyranyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi-6-alkylhalo, and OC0-6-alkylaryl; R7 is selected from Cl, Br, I, and -OCH3, and R5 is selected from C1-6-alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A.
In a still further embodiment, n is 1, 2, or 3; R4, R6, R8 and R9 are each H; R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC1-6-alkylhalo, and OCo-6-alkylaryl; R7 is selected from the group consisting of H, Cl, Br, I, C1-6-alkyl, and OC1-4-alkyl, and R5 is a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein the 5- to 7-membered ring is substituted by one or more A selected from the group consisting of Ci-6-alkyl-heterocyclyl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
In another embodiment, n is 1 ; R2, R3, R4, R6, R8 and R9 are each H; R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi-6-alkylhalo, and OC0-6-alkylaryl; R7 is selected from Cl, Br, I, and -OCH3, and R5 is selected from C1-6-alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A.
It will be understood by those of skill in the art that when compounds of the present invention contain one or more chiral centers, the compounds of the invention may exist in, and be isolated as, enantiomeric or diastereomeric forms, or as a racemic mixture. The present invention includes any possible enantiomers, diastereomers, racemates or mixtures thereof, of a compound of formula I. The optically active forms of the compound of the invention may be prepared, for example, by chiral chromatographic separation of a racemate, by synthesis from optically active starting materials or by asymmetric synthesis based on the procedures described thereafter.
It will also be appreciated by those of skill in the art that certain compounds of the present invention may exist as geometrical isomers, for example E and Z isomers of alkenes. The present invention includes any geometrical isomer of a compound of formula I. It will further be understood that the present invention encompasses tautomers of the compounds of formula I.
It will also be understood by those of skill in the art that certain compounds of the present invention may exist in solvated, for example hydrated, as well as unsolvated forms. It will further be understood that the present invention encompasses all such solvated forms of the compounds of formula I.
Within the scope of the invention are also salts of the compounds of formula I. Generally, pharmaceutically acceptable salts of compounds of the present invention are obtained using standard procedures well known in the art, for example, by reacting a sufficiently basic compound, for example an alkyl amine with a suitable acid, for example, HCl or acetic acid, to afford a physiologically acceptable anion. It is also possible to make a corresponding alkali metal (such as sodium, potassium, or lithium) or an alkaline earth metal (such as a calcium) salt by treating a compound of the present invention having a suitably acidic proton, such as a carboxylic acid or a phenol with one equivalent of an alkali metal or alkaline earth metal hydroxide or alkoxide (such as the ethoxide or methoxide), or a suitably basic organic amine (such as choline or meglumine) in an aqueous medium, followed by conventional purification techniques.
In one embodiment of the present invention, the compound of formula I may be converted to a pharmaceutically acceptable salt or solvate thereof, particularly, an acid addition salt such as a hydrochloride, hydrobromide, phosphate, acetate, fumarate, maleate, tartrate, citrate, methanesulphonate or j?-toluenesulphonate.
Specific examples of the present invention include the compounds described herein, their pharmaceutically acceptable salts, hydrates, solvates and optical isomers thereof. Pharmaceutical Compositions:
The compounds of the present invention may be formulated into conventional pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt or solvate thereof, in associaton with a pharmaceutically acceptable carrier or excipient. The pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories.
A solid carrier can be one or more substances, which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or table disintegrating agents. A solid carrier can also be an encapsulating material.
In powders, the carrier is a finely divided solid, which is in a mixture with the finely divided compound of the invention, or the active component. In tablets, the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
For preparing suppository compositions, a low-melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient sized moulds and allowed to cool and solidify. Suitable carriers include, but are not limited to, magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, low-melting wax, cocoa butter, and the like.
The term composition is also intended to include the formulation of the active component with encapsulating material as a carrier providing a capsule in which the active component (with or without other carriers) is surrounded by a carrier which is thus in association with it. Similarly, cachets are included.
Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration.
Liquid form compositions include solutions, suspensions, and emulsions. For example, sterile water or water propylene glycol solutions of the active compounds may be liquid preparations suitable for parenteral administration. Liquid compositions can also be formulated in solution in aqueous polyethylene glycol solution.
Aqueous solutions for oral administration can be prepared by dissolving the active component in water and adding suitable colorants, flavoring agents, stabilizers, and thickening agents as desired. Aqueous suspensions for oral use can be made by dispersing the finely divided active component in water together with a viscous material such as natural synthetic gums, resins, methyl cellulose, sodium carboxymethyl cellulose, and other suspending agents known to the pharmaceutical formulation art. Exemplary compositions intended for oral use may contain one or more coloring, sweetening, flavoring and/or preservative agents.
Depending on the mode of administration, the pharmaceutical composition will include from about 0.05%w (percent by weight) to about 99%w, more particularly, from about 0.10%w to 50%w, of the compound of the invention, all percentages by weight being based on the total weight of the composition.
A therapeutically effective amount for the practice of the present invention can be determined by one of ordinary skill in the art using known criteria including the age, weight and response of the individual patient, and interpreted within the context of the disease which is being treated or which is being prevented. Uses: We have discovered that the compounds of the present invention exhibit activity as pharmaceuticals, in particular as modulators of metabotropic glutamate receptors. More particularly, the compounds of the present invention exhibit activity as potentiators of the mGluR2 receptor. Such compounds and pharmaceutical compositions containing them are therefore useful in therapy, in particular for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction in an animal.
Neurological and psychiatric disorders amenable to treatment with compounds disclosed herein include, but are not limited to, disorders such as cerebral deficit subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, idiopathic and drug-induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, cerebral deficits secondary to prolonged status epilepticus, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, schizophrenia, anxiety (including generalized anxiety disorder, panic disorder, social phobia, obsessive compulsive disorder, and post-traumatic stress disorder (PTSD)), mood disorders (including depression, mania, bipolar disorders), circadian rhythm disorders (including jet lag and shift work), trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, inflammatory pain, and post-traumatic pain), tardive dyskinesia, sleep disorders (including narcolepsy), attention deficit/hyperactivity disorder, and conduct disorder.
The invention thus provides a use of any of the compounds according to formula I, or a pharmaceutically acceptable salt or solvate thereof, for the manufacture of a medicament for the treatment of any of the conditions discussed above.
Additionally, the invention provides a method for the treatment of a subject suffering from any of the conditions discussed above, whereby an effective amount of a compound according to formula I or a pharmaceutically acceptable salt or solvate thereof, is administered to a patient in need of such treatment. The invention also provides a compound of formula I or pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined for use in therapy.
In the context of the present specification, the term "therapy" also includes "prophylaxis" unless there are specific indications to the contrary. The term "therapeutic" and "therapeutically" should be construed accordingly. The term "therapy" within the context of the present invention further encompasses the administration of an effective amount of a compound of the present invention, to mitigate either a pre-existing disease state, acute or chronic, or to mitigate a recurring condition. This definition also encompasses prophylactic therapies for prevention of recurring conditions and continued therapy for chronic disorders.
In use for therapy in a warm-blooded animal such as a human, the compounds of the present invention may be administered in the form of a conventional pharmaceutical composition by any route including orally, intramuscularly, subcutaneously, topically, intranasally, intraperitoneally, intrathoracially, intravenously, epidurally, intrathecally, intracerebroventricularly and by injection into the joints. In particular embodiments of the invention, the route of administration is oral, intravenous, or intramuscular.
The dosage will depend on the route of administration, the severity of the disease, age and weight of the patient and other factors normally considered by the attending physician, who determines the individual regimen and dosage level for a particular patient.
As mentioned above, the compounds described herein may be provided or delivered in a form suitable for oral use, for example, in a tablet, lozenge, hard and soft capsule, aqueous solution, oily solution, emulsion, and suspension. Alternatively, the compounds may be formulated into a topical administration, for example, as a cream, ointment, gel, spray, or aqueous solution, oily solution, emulsion or suspension. The compounds described herein also may be provided in a form that is suitable for nasal administration, for example, as a nasal spray, nasal drops, or dry powder. The compounds can be administered to the vagina or rectum in the form of a suppository. The compounds described herein also may be administered parentally, for example, by intravenous, intravesicular, subcutaneous, or intramuscular injection or infusion. The compounds can be administered by insufflation (for example as a finely divided powder). The compounds may also be administered transdermally or sublingually. In addition to their use in therapeutic medicine, the compounds of formula I, or salts thereof, are useful as pharmacological tools in the development and standardisation of in vitro and in vivo test systems for the evaluation of the effects of inhibitors of mGluR-related activity in laboratory animals as part of the search for new therapeutics agents. Such animals include, for example, cats, dogs, rabbits, monkeys, rats and mice. Processes for Preparing:
The selection of a particular process to prepare a given compound is within the purview of the person of skill in the art. The choice of particular structural features and/or substituents may therefore influence the selection of one process over another.
Within these general guidelines, the following processes can be used to prepare exemplary subsets of compounds of this invention. Unless indicated otherwise, the variables described in the following schemes and processes have the same definitions as those given for formula I above.
In one process, for example, a compound of formula Ia:
Figure imgf000024_0001
is cyclized in the presence of an amine of the formula R'(CR8CR9)nNH2 to give a compound of formula Ib:
Figure imgf000024_0002
A compound of formula Ib is then cross-coupled with a suitable reagent containing R to yield a a compound according to formula Ic:
Figure imgf000024_0003
In one embodiment of this process, 5-substituted-7-methyl isoindolones are synthesized as depicted in Scheme 1 below. 4-bromo-2,6-dimethylaniline is converted to the corresponding nitrile under Sandmeyer reaction conditions. The nitrile is then hydrolyzed to the acid in a stepwise fashion. The amide can be obtained by basic hydrolysis. The amide is then diazotized and hydrolyzed with nitrososulphuric acid to provide the benzoic acid, which is subsequently protected as the methyl ester using standard conditions. The benzylic methyl group is monobrominated with N-bromosuccinimide using benzoyl peroxide as the radical initiator. This resultant intermediate is cyclized to the isoindolone with the appropriate amine in the presence of a base such as potassium carbonate. Finally, substituent R5 was introduced at C5 of the isoindolone using typical Buchwald, Suzuki or Stille cross-coupling reaction conditions and reagents. Scheme 1 :
Figure imgf000025_0001
(a) NaCN, CuCN, HCI; (b) NaOH; (c) nitrososulphuric acid; (d) MeI, K2CO3; (e) NBS, (PhCO2).,; (f) RICH2NH2, K2CO3; (g) R5H, BINAP, PdCI2(dppf), NaOtBu OR R5B(OH)2, PdCI2(dppf), K2CO3 OR R5SnBu3, Pd(PPh3)4
In another embodiment of this process, 5-substituted-7-chloro isoindolones are synthesized as depicted in Scheme 2 below. 4-bromo-2-methylbenzoic acid is chlorinated ortho to the acid using N-chlorosuccinimide and a palladium catalyst. In the manner analogous to that described above (Scheme 1), this acid was then esterified, brominated, and cyclized to yield the isoindolone intermediate. Substituent R5 is introduced similarly. Scheme 2:
Figure imgf000025_0002
(a) NCS, Pd(OAc)2; (b) MeI, K2CO3; (c) NBS, (PhCO2J2; (d) RICH2NH2, K2CO3;
(e) R5H, BINAP, PdCI2(dppf), NaOtBu OR R5B(OH)2, PdC!2(dppf), K2CO3 OR R5SnBu3, Pd(PPh3X, In yet another embodiment of this process, isoindolones that are substituted with an amide at C5 can be prepared as depicted in Scheme 3 below. Thus, an appropriately substituted 5-bromoisoindolone is converted to the corresponding nitrile using zinc cyanide in the presence of a palladium catalyst. The nitrile is then hydrolyzed under basic conditions to provide the benzoic acid, which was then coupled with various amines using methodologies that are well-known in the art to provide the final compounds. Scheme 3:
Figure imgf000026_0001
(a) Zn(CN)2, Pd(PPh3)4; (b) NaOH; (c) RR1NH, EDCI
Variations of the foregoing processes and additions thereto appear in the examples that follow. The person of ordinary skill in the art thus will appreciate that the compounds of this invention can be prepared by following or adapting one or more of the processes disclosed herein.
The invention is further illustrated by way of the following examples, which are intended to elaborate several embodiments of the invention. These examples are not intended to, nor are they to be construed to, limit the scope of the invention. It will be clear that the invention may be practiced otherwise than as particularly described herein. Numerous modifications and variations of the present invention are possible in view of the teachings herein and, therefore, are within the scope of the invention. General methods:
All starting materials are commercially available or earlier described in the literature.
^H and ^C NMR spectra were recorded either on Bruker 300, Bruker DPX400 or
Varian +400 spectrometers operating at 300, 400 and 400 MHz for 1H NMR respectively, using TMS or the residual solvent signal as reference, in deuterated chloroform as solvent unless otherwise indicated. All reported chemical shifts are in ppm on the delta-scale, and the fine splitting of the signals as appearing in the recordings (s: singlet, br s: broad singlet, d: doublet, t: triplet, q: quartet, m: multiplet).
Analytical in-line liquid chromatography separations followed by mass spectra detections, were recorded on a Waters LCMS consisting of an Alliance 2795 (LC) and a ZQ single quadropole mass spectrometer. The mass spectrometer was equipped with an electrospray ion source operated in a positive and/or negative ion mode. The ion spray voltage was ±3 kV and the mass spectrometer was scanned from m/z 100-700 at a scan time of 0.8 s. To the column, X-Terra MS, Waters, C8, 2.1 x 50mm, 3.5 mm, was applied a linear gradient from 5 % to 100% acetonitrile inlO mM ammonium acetate (aq.), or in 0.1% TFA
(aq.)-
Preparative reversed phase chromatography was run on a Gilson autopreparative HPLC with a diode array detector using an XTerra MS C8, 19x300mm, 7mm as column.
Purification by a chromatotron was performed on rotating silica gel / gypsum (Merck, 60 PF-254 with calcium sulphate) coated glass sheets, with coating layer of 1, 2, or 4 mm using a TC Research 7924T chromatotron.
Purification of products were also done using Chem Elut Extraction Columns (Varian, cat #1219-8002), Mega BE-SI (Bond Elut Silica) SPE Columns (Varian, cat # 12256018; 12256026; 12256034), or by flash chromatography in silica-filled glass columns. [0001] Microwave heating was performed in a Smith Synthesizer Single-mode microwave cavity producing continuous irradiation at 2450 MHz (Personal Chemistry AB, Uppsala, Sweden).
The pharmacological properties of the compounds of the invention can be analyzed using standard assays for functional activity. Examples of glutamate receptor assays are well known in the art as described in, for example, Aramori et ah, 1992, Neuron, 8:757; Tanabe et al, 1992, Neuron, 8:169; Miller etal, 1995, J. Neuroscience, 15:6103; Balazs, et al, 1997, J. Neurochemistry, 1997,69:151. The methodology described in these publications is incorporated herein by reference. Conveniently, the compounds of the invention can be studied by means of an assay that measures the mobilization of intracellular calcium, [Ca2+] j in cells expressing mGluR2.
Fluorometric Imaging Plate Reader (FLIPR) analysis was used to detect allosteric activators of mGluR2 via calcium mobilization. A clonal HEK 293 cell line expressing a chimeric mGluR2/CaR construct comprising the extracellular and transmembrane domains of human mGluR2 and the intracellular domain of the human calcium receptor, fused to the promiscuous chimeric protein Gαqj5 was used. Activation of this construct by agonists or allosteric activators resulted in stimulation of the PLC pathway and the subsequent mobilization of intracellular Ca2+ which was measured via FLIPR analysis. At 24-hours prior to analysis, the cells were trypsinized and plated in DMEM at 100,000 cells/well in black sided, clear-bottom, collagen I coated, 96-well plates. The plates were incubated under 5% CO2 at 37 0C overnight. Cells were loaded with 6μM fluo-3 acetoxymethylester (Molecular Probes, Eugene Oregon) for 60 minutes at room temperature. All assays were performed in a buffer containing 126 mM NaCl, 5 mM KCl, 1 mM MgCl2, 1 mM CaCl2, 20 mM Hepes, 0.06 μM DCG-IV (a Group II mGluR selective agonist), supplemented with 1.0 mg/ml D-glucose and 1.0 mg/ml BSA fraction IV (pH 7.4).
FLIPR experiments were done using a laser setting of 0.8 W and a 0.4 second CCD camera shutter speed. Extracellular fluo-3 was washed off and cells were maintained in 160 μL of buffer and placed in the FLIPR. An addition of test compound (0.01 μM to 30 μM in duplicate) was made after 10 seconds of baseline fluorescent readings were recorded on FLIPR. Fluorescent signals were then recorded for an additional 75 seconds at which point a second addition of DCG-IV (0.2 μM) was made and fluorescent signals were recorded for an additional 65 seconds. Fluorescent signals were measured as the peak height of the response within the sample period. Data was analyzed using Assay Explorer, and EC50 and Emax values (relative to maximum DCG-IV effect) were calculated using a four parameter logistic equation.
A [35S]-GTPyS binding assay was used to functionally assay mGluR2 receptor activation. The allosteric activator activity of compounds at the human mGluR2 receptor were measured using a [35S]-GTPyS binding assay with membranes prepared from CHO cells which stably express the human mGluR2. The assay is based upon the principle that agonists bind to G-protein coupled receptors to stimulate GDP-GTP exchange at the G-protein. Since [35S]-GTPyS is a non-hydrolyzable GTP analog, it can be used to provide an index of GDP-GTP exchange and, thus, receptor activation. The GTPyS binding assay therefore provides a quantitative measure of receptor activation. Membranes were prepared from CHO cells stably transfected with human mGluR2. Membranes (30 μg protein) were incubated with test compound (3 nM to 300 μM) for 15 minutes at room temperature prior to the addition of 1 μM glutamate, and incubated for 30 min at 30 0C in 500 μL assay buffer (20 mM HEPES, 100 mM NaCl, 10 raM MgCl2), containing 30 μM GDP and 0.1 nM [35S]-GTPyS (1250 Ci/mmol). Reactions were carried out in triplicate in 2 mL polypropylene 96-well plates. Reactions were terminated by vacuum filtration using a Packard 96-well harvester and Unifilter-96, GF/B filter microplates. The filter plates were washed 4 x 1.5 mL with ice-cold wash buffer (10 mM sodium phosphate buffer, pH 7.4). The filter plates were dried and 35 μL of scintillation fluid (Microscint 20) was added to each well. The amount of radioactivity bound was determined by counting plates on the Packard TopCount. Data was analyzed using GraphPad Prism, and EC50 and Emax values (relative to the maximum glutamate effect) were calculated using non-linear regression.
Generally, the compounds of the present invention were active in assays described herein at concentrations (or with EC50 values) less than 10 μM. Exemplary Processes: Compound 1 : 2-[4-(4-Fluoro-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one
Figure imgf000029_0001
A mixture of 2-bromomethyl-benzoic acid methyl ester (0.108 g, 0.5 mmol), 4-(4-fluoro-phenoxy)-benzylamine (0.115 g, 0.5 mmol), and K2CO3 (0.235 g, 1.7 mmol) in toluene (5 mL) was heated with stirring at 100 0C for 2 h. Workup and silica gel column chromatography using 30% ethyl acetate in hexane afforded
2-[4-(4-fluoro-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one (0.100 g, 60%). 1H NMR (300 MHz, CDCl3): δ (ppm) 4.29 (s, 2H), 4.78 (s, 2H) 6.83 - 7.56 (m, 1 IH), 7.89 (d, IH). GC-MS: m/z 333 (M)+. Compound 2: 2-[4-(4-Trifluoromethyl-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one
Figure imgf000030_0001
A mixture of 2-bromomethyl-benzoic acid methyl ester (0.115 g, 0.5 mmol), 4-(4-trifluoromethyl-phenoxy)-benzylamine (0.133 g, 0.5 mmol), and K2CO3 (0.235 g, 1.7 mmol) in toluene (5 mL) was heated with stirring at 100 °C for 2 h. Workup and silica gel column chromatography using 30% ethyl acetate in hexane afforded 2-[4-(4-trifluoromethyl-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one (0.116 g, 60%). 1H NMR (300 MHz, CDCl3): δ (ppm) 4.32 (s, 2H), 4.81 (s, 2H) 7.01 - 7.56 (m, 1 IH), 7.91 (d, IH). GC-MS: m/z 383 (M)+.
Compound 3 : - { 3 - [4-(4-Fluoro-phenoxy)-phenyl] -propyl} -7-iodo-2,3 -dihydro-isoindol- 1 -one
Figure imgf000030_0002
A mixture of 2-bromomethyl-6-iodo-benzoic acid methyl ester (0.245 g, 0.7 mmol), 3-[4-(4-fluoro-phenoxy) phenyl] -propylamine ( 0.193 g, 0.8 mmol), and K2CO3 (0.207 g, 1.5 mmol) in toluene (5 mL) was heated with stirring at 100 °C for 2 h. Workup and silica gel column chromatography using 30% ethyl acetate in hexane afforded 2-{3-[4-(4-fluoro-phenoxy)-phenyl]-propyl}-7-iodo-2,3-dihydro-isoindol-l-one (0.086 g, 25%). 1H NMR (300 MHz, CDCl3): δ (ppm) 2.01 (m, 2H), 2.65 (t, 2H), 3.67 (t, 2H), 4.26 (s, 2H), 6.84 - 7.42 (m, 10H), 7.90 (d, IH). GC-MS: m/z 487 (M)+. Compound 4: 7-Iodo-2-[3-(2-methoxy-phenoxy)-benzyl]-2,3-dihydro-isoindol- 1 -one
Figure imgf000030_0003
A mixture of 2-bromomethyl-6-iodo-benzoic acid methyl ester (0.178 g, 0.5 mmol), 3-(2-methoxy-phenoxy)-benzylamine ( 0.160 g, 0.6 mmol), and K2CO3 (0.138 g, 1.0 mmol) in toluene (5 mL) was heated with stirring at 100 °C for 2 h. Workup and silica gel column chromatography using 30% ethyl acetate in hexane afforded 7-iodo-2-[3-(2-methoxy-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one (0.056 g, 24%). 1H NMR (300 MHz, CDCl3): δ (ppm) 3.82 (s, 3H), 4.16 (s, 2H), 4.74 (s, 2H), 6.84 -7.40 (m, 10H), 7.92 (d, IH). GC-MS: m/z 471 (M)+.
The following Examples were prepared by processes analogous to those described in Compounds 1, 2, 3 and 4: Example 1 : 5-Bromo-2-[4-(2-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one
Figure imgf000031_0001
0.80 g, 42 %, yellow oil. 7.37 (d, 2H), 7.27 (d, 2H), 7.02-7.22 (m, 4H), 6.95 (d, 2H), 4.73 (s,
2H), 4.21 (s, 2H), 2.74 (s, 3H).
Example 2: 5-Bromo-2-[4-(3-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one
Figure imgf000031_0002
0.58 g, 26 %, yellow solid. 7.38 (d, 2H), 7.27-7.33 (m, 3H), 7.02 (d, 2H), 6.75-6.85 (m, 2H),
6.66-6.72 (m, IH), 4.76 (s, 2H), 4.24 (s, 2H), 2.75 (s, 3H).
Example 3: 5-Bromo-2-[4-(4-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one
Figure imgf000032_0001
0.38 g, 34 %, yellow oil. 7.37 (d, 2H), 7.27 (d, 2H), 6.90-7.10 (m, 6H)5 4.73 (s, 2H), 4.22 (s,
2H), 2.75 (s, 3H).
Example 4: 5-Bromo-7-methyl-2-[4-(pyridin-2-yloxy)benzyl]-2,3-dihydro-isoindol- 1 -one
Figure imgf000032_0002
0.42 g, 40 %, yellow oil. 8.17-8.22 (m, IH), 7.67-7.75 (m, IH), 7.33-7.39 (m, 4H), 7.13 (d,
2H), 7.00-7.05 (m, IH), 6.93 (d, 2H), 4.77 (s, 2H), 4.25 (s, 2H), 2.75 (s, 3H).
Example 5 : 5-Bromo-7-methyl-2-[4-(pyridin~3-yloxy)benzyl]-2,3-dihydro-isoindol- 1 -one
Figure imgf000032_0003
0.19 g, 9 %, orange solid. 8.37-8.42 (m, 2H), 7.38 (d, 2H), 7.28-7.33 (m, 4H), 7.01 (d, 2H), 4.76 (s, 2H), 4.24 (s, 2H), 2.75 (s, 3H). Example 6: 2-[4-(2-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-2-yl-2,3-dihydro-isoindol-l-one
Figure imgf000033_0001
64 mg, 64 %, pale yellow solid. 8.70-8.75 (m, IH), 7.77-7.85 (m, 4H), 7.27-7.32 (m, 3H), 7.02-7.23 (m, 4H), 6.97 (d, 2H), 4.79 (s, 2H), 4.31 (s, 2H), 2.85 (s, 3H). Example 7 : 2-[4-(2-Fluoro-phenoxy)-benzyl] -7-methyl-5 -pyridin-3 -yl-2,3 -dihydro -isoindol-1-one
Figure imgf000033_0002
38 mg, 38 %, pale yellow oil. 8.86 (d, IH), 8.65 (dd, IH), 7.85-7.92 (m, IH), 7.38-7.41 (m, 3H), 7.30 (d, 2H), 7.02-7.23 (m, 4H), 6.96 (d, 2H), 4.79 (s, 2H), 4.32 (s, 2H), 2.85 (s, 3H). Example 8: 2-[4-(2-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyrazin-2-yl-2,3-dihydro -isoindol-1-one
Figure imgf000033_0003
24 mg, 24 %, off-white solid. 9.07 (d, IH), 8.68 (dd, IH), 8.58 (d, IH), 7.87 (s, 2H), 7.29 (d, 2H), 7.02-7.23 (m, 4H), 6.96 (d, 2H), 4.79 (s, 2H), 4.34 (s, 2H), 2.87 (s, 3H). Example 9: 7-Methyl-5-pyrazin-2-yl-2-[4-(pyridin-2-yloxy)-benzyl]-2,3-dihydro -isoindol-1-one
Figure imgf000034_0001
22 mg, 22 %, off-white solid. 9.08 (d, IH), 8.67-8.69 (m, IH)5 8.58 (d, IH), 8.15-8.25 (m,
IH), 7.87 (s, 2H), 7.65-7.75 (m, IH), 7.39 (d, 2H), 7.14 (d, 2H), 6.98-7.05 (m, IH), 6.93 (d,
IH), 4.83 (s, 2H), 4.38 (s, 2H), 2.88 (s, 3H).
Example 10: 2-[4-(4-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-2-yl-2,3-dihydro
-isoindol-1-one
Figure imgf000034_0002
43 mg, 51 %, yellow oil. 8.72-8.74 (m, IH), 7.77-7.86 (m, 4H), 7.28-7.32 (m, 3H), 6.80-7.04
(m, 6H), 4.78 (s, 2H), 4.32 (s, 2H), 2.86 (s, 3H).
Example 11 : 2-[4-(4-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-3-yl-2,3-dihydro
-isoindol-1-one
Figure imgf000034_0003
22 mg, 25 %, yellow oil. 8.86 (d, IH), 8.63-8.66 (m, IH), 7.85-7.95 (m, IH), 7.40-7.43 (m, 3H), 7.30 (d, 2H), 6.90-7.05 (m, 6H), 4.79 (s, 2H), 4.33 (s, 2H), 2.85 (s, 3H). Example 12: 2-[4-(4-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyrazin-2-yl-2,3-dihydro -isoindol-1-one
Figure imgf000035_0001
21 mg, 25 %, pale yellow solid. 9.07 (d, IH), 8.67-8.69 (m, IH)3 8.58 (d, IH), 7.87 (s, 2H),
7.31 (d, 2H), 6.90-7.08 (m, 6H), 4.79 (s, 2H), 4.34 (s, 2H), 2.87 (s, 3H).
Example 13 : 2-[4-(3-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-2-yl-2,3-dihydro
-isoindol-1-one
Figure imgf000035_0002
41 mg, 41 %, colourless oil. 8.70-8.75 (m, IH), 7.75-7.88 (m, 4H), 7.25-7.37 (m, 4H), 7.03 (d, 2H), 6.75-6.85 (m, 2H), 6.65-6.73 (m, IH), 4.81 (s, 2H), 4.14 (s, 2H), 2.86 (s, 3H). Example 14: 2-[4-(3-Fluoro-phenoxy)-benzyl]-7-methyl-5-pyrazin-2-yl-2,3-dihydro -isoindol-1-one
Figure imgf000035_0003
27 mg, 27 %, off-white solid. 9.08 (d, IH), 8.67-8.69 (m, IH), 8.58 (d, IH), 7.87-7.89 (m,
2H), 7.35 (d, 2H), 7.27-7.29 (m, IH), 7.03 (d, 2H), 6.75-6.83 (m, 2H), 6.68-6.72 (m, IH),
4.81 (s, 2H), 4.36 (s, 2H), 2.88 (s, 3H).
Example 15: 2-[4-(2-Fluoro-phenoxy)-benzyl]-7-methyl-5-thiazol-2-yl-2,3-dihydro
-isoindol-1-one
Figure imgf000036_0001
23 mg, 26 %, yellow oil. 7.92 (d, IH), 7.84 (s, IH), 7.82 (s, IH)5 7.41 (d, IH), 7.30 (d, 2H), 7.03-7.25 (m, 4H), 6.96 (d, 2H), 4.77 (s, 2H), 4.30 (s, 2H), 2.84 (s, 3H).

Claims

WHAT IS CLAIMED IS:
1. A compound according to formula I:
Figure imgf000037_0001
wherein:
R1 is a 3- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said ring is substituted by one or more B;
R2 and R3 are independently selected from the group consisting of H and Ci-6-alkyl;
R4 is H and R6 is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, Ci-6-alkyl, and OCi-6alkyl;
R5 is selected from the group consisting of H, F, Cl, Br, I, nitro, CN, Ci-6-alkyl, OC0-6-alkyl, d-6-alkylhalo, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OCo-e-alkyl-C^s-cycloalkyl, aryl, Ci-6-alkylaryl, Ci_6-alkylheteroaryl, OCi-6-alkylaryl, OCi-6-alkylheteroaryl, Ci-e-alkylheterocycloalkyl, Oheterocycloalkyl, OCi-β-alkylheterocycloalkyl, C(O)H, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, 0(CN)OR10, C1-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, C0-6-alkylCO2R10, Ci-6-alkylcyano, OC2-6-alkylcyano, Co-6-alkylNRloRn, OC^-alkylNR'V1, Ci-6-alkyl(CO)NR10Rπ, OC1-6-^yI(CO)NR10R1 \ C0-6-alkylNR10(CO)R1 ' , OC2-6-alkylNR10(CO)R1 ' , C0-6-alkylNR10(CO)NR10Rn, C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, Co-e-alky^SO^NR^R11, OC2-6-alkyl(SO2)NR10Rn, Co-6-alkylNR10(S02)R1 ', OC2-6-alkylNR10(SO2)Rn, C0-6-alkylNR10(SO2)NR10R11, OC2-6-alkylNR10(SO2)NR10R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, Co-6-alkylNR10(CO)ORπ, OC2-6-BIlCyINR1 ^CO)OR1 \ SO3R10, and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, OCi-4-alkyl and Ci-6-alkyl;
R and R are independently selected from the group consisting of H, F, Cl, Br, I, nitro, cyano, Ci-6-alkyl, Ci-6-alkylhalo, OC1-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, and OC2-6-alkynyl, or, where n is greater than 1 , two or more R8 and/or R9 on adjacent carbon atoms may be absent to form an alkenyl or alkynyl moiety;
R10 and R11 are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, Ci-6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, d-ό-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, Ci_6-alkylaryl, OC0-6-alkylaryl, Co-o-alkyl-heterocycloalkyl, OC^-alkyl-heterocycloalkyl, heteroaryl, and C1-6alkylheteroaryl, wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S and any cyclic moiety is optionally substituted with a substituent selected from alkyl, halo, hydroxyl, Oalkyl, haloalkyl and Ohaloalkyl;
A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, Ci-6-alkyl, Ci-6-alkylhalo, OCi-6alkyl, OC]-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci-β-alkyl-Cs-s-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, OC0-6-alkylaryl, Ci-o-alkyl-heterocyclyl, Ci-e-alkyl-heterocycloalkyl, OC^-alkyl-heterocycloalkyl, (CO)R10, O(CO)R10, 0(CO)OR10, O(CNR10)ORπ, Ci-6-alkylOR10, OC2-6-alkylOR10, Ci-6-alkyl(CO)R10, OCi-6-alkyl(CO)R10, C0-6-alkylCO2R10 5 OC1-6-alkylCO2R10, Ci-6-alkylcyano, OC2-6-alkylcyano, C0-6-alkylNR10Rπ, OC2-6-alkylNR10R11, Co-6-alkyl(CO)NR10R11, OC1-6-alkyl(CO)NR10R11, Co-6-alkylNR10(CO)R11, OC2-6-alkylNR10(CO)R11, C0-6-alkylNR10(CO)NR10R11, Co-e-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C1-6-alkylSO2R10,
Figure imgf000038_0001
Co-6-alkylNR10(S02)R1 \ OC2-6-alkylNR10(SO2)R1 \ Co-6-alkylNR10(S02)NR10R1 ' , OC2-6-alkylNR10(SO2)NR10R11, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, Co-6-alkylNR10(CO)ORπ, OC2-6-alkylNR10(CO)OR11, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said 5- to 7-membered ring is optionally substituted by one or more of R10 and R11;
B is selected from the group consisting of Cs-s-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, C0-6-alkylaryl, OC0-6-alkylaryl, Ci-δ-alkyl-heterocycloalkyl, Ci-β-alkyl-heterocycloalkyl, OCo-e-alkyl-heterocycloalkyl, Co-6-alkyl-heteroaryl and OC0-6-alkyl-heteroaryl, wherein any cyclic moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO2R, SO2R and CN; and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof, with a proviso that said compound is not: 2- [4-(4-fluoro-phenoxy)-benzyl] -2 ,3 -dihydro-isoindol- 1 -one ; 2-[4-(4-trifluoromethyl-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one; 2- { 3 - [4-(4-fluoro-phenoxy)-phenyl] -propyl } -7-iodo-2,3 -dihydro-isoindol- 1 -one, or 7-iodo-2-[3-(2-methoxy-phenoxy)-benzyl]-2,3-dihydro-isoindol-l-one.
2. A compound according to Claim 1 , wherein:
R1 is phenyl wherein said phenyl is substituted by one or more B; R2 and R3 are independently selected from the group consisting of H, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, heterocycloalkyl, C3-8-cycloalkyl, Ci-6-alkyl-aryl, C1-6-alkyl-heteroaryl, Ci^-alkyl-heterocycloalkyl, and Ci-6-alkyl-C3-8-cycloalkyl, wherein R2 and R3 may be substituted by one or more A;
R4 and R6 are independently selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, C1-6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OCo-e-alkyl-Cs-s-cycloalkyl, aryl, C1-6-alkylaryl, OC0-6-alkylaryl, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, 0(CNR10PR1 ', C1-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, C0-6-alkylCO2R10 3 OC1-6-alkylCO2R10, CI-6-alkylcyano, OC2-6-alkylcyano, Co-6-alkylNRloRπ, OC2-6-alkylNRI0Rπ, Ci-6-alkyl(CO)NRI0RI 1, OCi-6-alkyl(CO)NR10R11, C0-6-alkylNR10(CO)R11, OC2.6-alkylNRI0(CO)R11, Cce-alkylNR1 ^CO)NR10R11, C0-6-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, Co-6-alkyl(S02)NR10R1 I, OC2-6-alkyl(S02)NR10R11, Co-6-alkylNR10(S02)R11, OC2-6-alkylNR10(SO2)R' ' , Co-β-alkylNR1 ^SO2)NR10R1 ', OC2-6-alkylNR10(SO2)NRI0R1 ' , (CO)NR10R11, 0(CO)NR10R1^ NR10OR11, Cce-alkylNR1 ^CO)OR1 ', OC2-6-alkylNR10(CO)ORn, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R4 and R may be substituted by one or more A, and wherein any cycloalkyl ior aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S;
R5 is selected from the group consisting of H, F, Cl5 Br, I, nitro, CN, C1-6-alkyl, OCo-6-alkyl, C1-6-alkylhalo, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2.6-alkynyl, OC2-6-alkynyl, C3-s-cycloalkyl, C1-6-alkyl-C3-8-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, Ci_6-alkylheteroaryl, OCi-6-alkylaryl, OCi-6-alkylheteroaryl, Ci-ό-alkylheterocycloalkyl, Oheterocycloalkyl, OCj-e-alkylheterocycloalkyl, C(O)H, (CO)R10, 0(CO)R10, 0(CO)OR10, C(O)OR10, 0(CN)OR10, C1-6-alkylOR10, OC2-6-alkylOR10, C1-6-alkyl(CO)R10, OC1-6-alkyl(CO)R10, C0.6-alkylCO2R10, C1-6-alkylcyano, OC2-6-alkylcyano, Co-6-alkylNRloRπ, OC2-6-alkylNR10R11, C1-6-alkyl(CO)NR10Rπ, OC1-6-^yI(CO)NR10R11, C0-6-alkylNR10(CO)R11, OC2-6-alkylNR10(CO)R11, Co-6-alkylNR10(CO)NR10R11, C0-6-alkylSR10, OC2-6-alkylSR10, Co-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C0-6-alkylSO2R10, OC2-6-alkylSO2R10, Co-e-alky^SO^NR^R11, OC^e-alkyKSO^NR^R1 \ C0-6-alkylNR10(SO2)R1 \ OC2-6-alkylNR10(SO2)R1 ', C0-6-alkylNR10(SO2)NR10R11, OC2-6-alkylNR10(SO2)NR10Rπ, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNRI0(CO)ORn, OC2-6-alkylNR' ^CO)OR1 ', SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S; R7 is selected from the group consisting of H, F, Cl, Br, I, nitro, cyano,
Figure imgf000041_0001
Ci-6-alkyl, Ci-6-alkylhalo, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, and C3-8-cycloalkyl;
R8 and R9 are both H;
R10 and R11 are independently selected from the group consisting of H, hydroxy, oxo, F, Cl, Br, I, nitro, cyano, d.6-alkyl, Ci-6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-CyClOaIlCyI, Ci-6-alkyl-C3-8-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, OC0-6-alkylaryl, Co-e-alkyl-heterocycloalkyl, OCi-β-alkyl-heterocycloalkyl, heteroaryl, and C1-6alkylheteroaryl, wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S and any cyclic moiety is optionally substituted with a substituent selected from alkyl, halo, hydroxyl, Oalkyl, haloalkyl and Ohaloalkyl;
A is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, oxo, C1-6-alkyl, C1-6-alkylhalo, OCi-6alkyl, OCi-6-alkylhalo, C2-6-alkenyl, OC2-6-alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, Ci.ό-alkyl-Cs-s-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, OC0-6-alkylaryl, Ci-ό-alkyl-heterocyclyl, Ci-e-alkyl-heterocycloalkyl, OCo^-alkyl-heterocycloalkyl, (CO)R10, 0(CO)R10, 0(CO)OR10, O(CNR10)ORn, Ci-6-alkylOR10, OC2-6-alkylOR10,
Figure imgf000041_0002
C0-6-alkylCO2R10, OC1-6-alkylCO2R10, Ci-6-alkylcyano, OC2-6-alkylcyano, C0-6-alkylNR10Rn, OC2-6-^yINR10R1 \ C0-6-alkyl(CO)NR10R1 ', OCi-6-alkyl(CO)NR10Rπ, C0-6-alkylNR10(CO)R11, OC2-6-alkylNR10(CO)R11, C0-6-alkylNR10(CO)NR10R11, Co-e-alkylSR10, OC2-6-alkylSR10, C0-6-alkyl(SO)R10, OC2-6-alkyl(SO)R10, C]-6-alkylSO2R10, OC^e-alkylSO^^^o-e-alkyKSO^NR^R11, OC2-6-alkyl(SO2)NR10Rπ, C0-6-alkylNR10(SO2)R1 \ OC2-6-alkylNR10(SO2)R' \ C0-6-alkylNR10(SO2)NR10R1 ', OC2-6-alkylNR10(SO2)NR10RH, (CO)NR10R11, 0(CO)NR10R11, NR10OR11, C0-6-alkylNR10(CO)OR11, OC2-6-alkylNR10(CO)ORn, SO3R10 and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein said 5- to 7-membered ring is optionally substituted by one or more of R10 and R11; B is selected from the group consisting of C0-6-alkylaryl and OCo-6-alkylaryl, wherein any aryl moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo, hydroxy, alkoxy, oxo, COR, CO2R, SO2R and CN; and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
3. A compound according to Claim 1, wherein:
R1 is phenyl wherein said phenyl is substituted by one or more B;
R2 and R3 are independently selected from the group consisting of H and C]-6-alkyl;
R4 is H and R6 is selected from the group consisting of H, hydroxy, F, Cl, Br, I, nitro, cyano, C1-6-alkyl, and OC1-6alkyl;
R5 is selected from the group consisting of H, F, Cl, Br, I, or a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S;
R7 is selected from the group consisting of H or C1-6-alkyl;
R8 and R9 are both H;
B is OCo-6-alkylaryl, wherein said aryl moiety is substituted with at least one substituent selected from the group consisting of halo, alkyl, alkylhalo and alkoxy, and n is 1; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
4. A compound according to Claim 1 , wherein: n is 1, 2, or 3 and when n is greater than 1, two or more of R8 and R9 on adjacent carbon atoms can be missing so as to form partially or fully unsaturated moieties; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
5. A compound according to Claim 1, wherein: n is 1 and R8 and R9 are each H; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
6. A compound according to Claim 1, wherein: R4 and R6 are each H; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
7. A compound according to Claim 1, wherein:
R1 is a 5- to 7-membered ring selected from the group consisting of aryl, C3-8-cycloalkyl, cycloalkenyl, and heterocyclyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi-6-alkylhalo, and OC0-6-alkylaryl; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
8. A compound according to Claim 1, wherein:
R1 is selected from phenyl, naphthyl, C3-8-cycloalkyl, cycloalkenyl, furanyl, tetrahydrofuranyl, thiophenyl, pyridyl, oxadiazolyl, quinolinyl, piperazinyl, and tetrahy dropyr any 1 ; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
9. A compound according to Claim 1 , wherein:
R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC1-6-alkylhalo, and OCo-6-alkylaryl; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
10. A compound according to Claim 9, wherein:
R2, R3, R4, R6, R8, and R9 are each H and n is 1, and
R7 is selected from H, Cl, Br, I, Ci-6-alkyl, and OC1-4-alkyl; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
11. A compound according to Claim 1 , wherein:
R5 is selected from the group consisting of C3-8-cycloalkyl, Ci-6-alkyl-C3-8-cycloalkyl, OCo-6-alkyl-C3-8-cycloalkyl, aryl, C1-6-alkylaryl, OCi-6-alkylaryl, and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S; wherein R5 may be substituted by one or more A, and any cycloalkyl or aryl is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
12. A compound according to Claim 11 , wherein:
R5 is selected from Ci-6-alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
13. A compound according to Claim 12, wherein:
R5 is a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, which ring is substituted by one or more A selected from the group consisting of Ci-6-alkyl-heterocyclyl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
14. A compound according to Claim 1, wherein: n is 1, 2, or 3;
R4, R6, R8 and R9 are each H;
R1 is selected from the group consisting of aryl, C3-8-cycloalkyl, cycloalkenyl, and heterocyclyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi-6-alkylhalo, and OCo-6-alkylaryl;
R7 is selected from the group consisting of H, Cl, Br, I, Ci-6-alkyl, and OCi-4-alkyl, and
R5 is selected from the group consisting of C3-8-cycloalkyl, C]-6-alkyl-C3-8-cycloalkyl, OC0-6-alkyl-C3-8-cycloalkyl, aryl, Ci-6-alkylaryl, OC]-6-alkylaryl, and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A, and wherein any cyclic moiety is optionally fused to a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of C, N, O and S; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
15. A compound according to Claim 1, wherein: n is 1, 2, or 3;
R4, R6, R8 and R9 are each H;
R1 is selected from phenyl, naphthyl, C3-8-cycloalkyl, cycloalkenyl, furanyl, tetrahydrofuranyl, thiophenyl, pyridyl, oxadiazolyl, quinolinyl, piperazinyl, and tetrahydropyranyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC].6-alkylhalo, and OC0-6-alkylaryl; R7 is selected from Cl, Br, I, and - OCH3, and R5 is selected from Ci-6-alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
16. A compound according to Claim 1, wherein: n is 1, 2, or 3;
R4, R6, R8 and R9 are each H;
R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OC1-6-alkylhalo, and OC0-6-alkylaryl; R7 is selected from the group consisting of H, Cl, Br, I, C1-6-alkyl, and OC1-4-alkyl, and R5 is a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein the 5- to 7-membered ring is substituted by one or more A selected from the group consisting of Ci-6-alkyl-heterocyclyl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
17. A compound according to Claim 1, wherein: n is 1;
R2, R3, R4, R6, R8 and R9 are each H;
R1 is phenyl optionally substituted by one or more A selected from the group consisting of F, Cl, Br, I, OCi-6-alkylhalo, and OC0-6-alkylaryl; R7 is selected from Cl, Br, I, and -OCH3, and R5 is selected from C1-6-alkylaryl and a 5- to 7-membered ring that may contain one or more heteroatoms independently selected from the group consisting of N, O and S, wherein R5 may be substituted by one or more A; or a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
18. A compound according to Claim 1 , selected from: 5-bromo-2-[4-(2-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one; 5-bromo-2-[4-(3-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one; 5-bromo-2-[4-(4-fluorophenoxy)benzyl]-7-methyl-2,3-dihydro-isoindol-l-one; 5-bromo-7-methyl-2-[4-(pyridin-2-yloxy)benzyl]-2,3-dihydro-isoindol-l-one;
5-bromo-7-methyl-2-[4-(pyridin-3-yloxy)benzyl]-2,3-dihydro-isoindol-l-one;
2-[4-(2-fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-2-yl-2,3-dihydro-isoindol-l-one;
2-[4-(2-fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-3-yl-2,3-dihydro-isoindol-l-one;
2-[4-(2-fluoro-phenoxy)-benzyl]-7-methyl-5-pyrazin-2-yl-2,3-dihydro-isoindol-l-one;
7-methyl-5-pyrazin-2-yl-2-[4-(pyridin-2-yloxy)-benzyl]-2,3-dihydro-isoindol-l-one;
2-[4-(4-fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-2-yl-2,3-dihydro-isoindol-l-one;
2-[4-(4-fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-3-yl-2,3-dihydro-isoindol-l-one;
2-[4-(4-fluoro-phenoxy)-benzyl]-7-methyl-5-pyrazin-2-yl-2,3-dihydro-isoindol-l-one;
2-[4-(3-fluoro-phenoxy)-benzyl]-7-methyl-5-pyridin-2-yl-2,3-dihydro-isoindol-l-one;
2-[4-(3-fluoro-phenoxy)-benzyl]-7-methyl-5-pyrazin-2-yl-2,3-dihydro-isoindol-l-one, and
2-[4-(2-fluoro-phenoxy)-benzyl]-7-methyl-5-thiazol-2-yl-2,3-dihydro-isoindol-l-one.
19. A pharmaceutical composition comprising a compound according to any one of claims 1 - 18 and a pharmaceutically acceptable carrier or excipient.
20. A compound according to any one of claims 1 - 18 for use as a medicament.
21. The use of a compound according to any one of claims 1 - 18 in the manufacture of a medicament for the therapy of neurological and psychiatric disorders associated with glutamate dysfunction.
22. The use of claim 21, wherein the neurological and psychiatric disorders are selected from cerebral deficit subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia, AIDS-induced dementia, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, idiopathic and drug-induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, cerebral deficits secondary to prolonged status epilepticus, migraine, migraine headache, urinary incontinence, substance tolerance, substance withdrawal, psychosis, schizophrenia, anxiety, generalized anxiety disorder, panic disorder, social phobia, obsessive compulsive disorder, and post-traumatic stress disorder (PTSD), mood disorders, depression, mania, bipolar disorders, circadian rhythm disorders, jet lag, shift work, trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain, acute pain, chronic pain, severe pain, intractable pain, neuropathic pain, inflammatory pain, and post-traumatic pain, tardive dyskinesia, sleep disorders, narcolepsy, attention defϊcit/hyperactivity disorder, and conduct disorder.
23. A method for the treatment or prevention of neurological and psychiatric disorders associated with glutamate dysfunction in an animal in need of such treatment, comprising the step of administering to said animal a therapeutically effective amount of a compound according to any one of claims 1 - 18.
24. A method for the treatment or prevention of neurological and psychiatric disorders associated with glutamate dysfunction in an animal in need of such treatment, comprising the step of administering to said animal a therapeutically effective amount of a pharmaceutical composition according to claim 23.
25. The method according to claim 23 or 24, wherein the neurological and psychiatric disorders are selected from cerebral deficit subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia, AIDS-induced dementia, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, idiopathic and drug-induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, cerebral deficits secondary to prolonged status epilepticus, migraine, migraine headache, urinary incontinence, substance tolerance, substance withdrawal, psychosis, schizophrenia, anxiety, generalized anxiety disorder, panic disorder, social phobia, obsessive compulsive disorder, and post-traumatic stress disorder (PTSD), mood disorders, depression, mania, bipolar disorders, circadian rhythm disorders, jet lag, shift work, trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain, acute pain, chronic pain, severe pain, intractable pain, neuropathic pain, inflammatory pain, and post-traumatic pain, tardive dyskinesia, sleep disorders, narcolepsy, attention defϊcit/hyperactivity disorder, and conduct disorder.
26. The method according to claim 25, wherein the neurological and psychiatric disorders are selected from Alzheimer's disease, cerebral deficits secondary to prolonged status epilepticus, substance tolerance, substance withdrawal, psychosis, schizophrenia, anxiety, generalized anxiety disorder, panic disorder, social phobia, obsessive compulsive disorder, and post-traumatic stress disorder (PTSD), mood disorders, depression, mania, and bipolar disorders.
27. A process for the preparation of a compound of formula Ic comprising: (a) cyclizing a compound of the formula Ia:
Figure imgf000049_0001
in the presence of an amine of the formula Rl(CR8R9)nNH2 into a compound of the formula
Ib:
Figure imgf000049_0002
(b) cross-coupling a compound of formula Ib with a reagent comprising R5 to yield a compound according to formula Ic:
Figure imgf000049_0003
wherein the variables are defined according to claim 1.
PCT/US2006/005246 2005-08-12 2006-02-15 Metabotropic glutamate-receptor-potentiating isoindolones WO2007021308A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2008525976A JP5031745B2 (en) 2005-08-12 2006-02-15 Metaindole glutamate receptor potentiating isoindolone
US12/063,007 US7807706B2 (en) 2005-08-12 2006-02-15 Metabotropic glutamate-receptor-potentiating isoindolones
EP06720758A EP1912939A1 (en) 2005-08-12 2006-02-15 Metabotropic glutamate-receptor-potentiating isoindolones
US12/861,336 US8153638B2 (en) 2005-08-12 2010-08-23 Metabotropic glutamate-receptor-potentiating isoindolones

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/US2005/028760 WO2006020879A1 (en) 2004-08-13 2005-08-12 Isoindolone compounds and their use as metabotropic glutamate receptor potentiators
USPCT/US2005/028760 2005-08-12

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/028760 Continuation WO2006020879A1 (en) 2004-08-13 2005-08-12 Isoindolone compounds and their use as metabotropic glutamate receptor potentiators

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/861,336 Continuation US8153638B2 (en) 2005-08-12 2010-08-23 Metabotropic glutamate-receptor-potentiating isoindolones

Publications (1)

Publication Number Publication Date
WO2007021308A1 true WO2007021308A1 (en) 2007-02-22

Family

ID=36464390

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2006/005247 WO2007021309A1 (en) 2005-08-12 2006-02-15 Substituted isoindolones and their use as metabotropic glutamate receptor potentiators
PCT/US2006/005246 WO2007021308A1 (en) 2005-08-12 2006-02-15 Metabotropic glutamate-receptor-potentiating isoindolones

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US2006/005247 WO2007021309A1 (en) 2005-08-12 2006-02-15 Substituted isoindolones and their use as metabotropic glutamate receptor potentiators

Country Status (4)

Country Link
EP (2) EP1912939A1 (en)
JP (2) JP5031745B2 (en)
CN (2) CN101309905A (en)
WO (2) WO2007021309A1 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008130853A1 (en) * 2007-04-17 2008-10-30 Astrazeneca Ab Hydrazides and their use as metabotropic glutamate receptor potentiators - 681
WO2008150232A1 (en) * 2007-06-07 2008-12-11 Astrazeneca Ab Metabotropic glutamate receptor oxadiazole ligands and their use as potentiators - 841
WO2008150233A1 (en) * 2007-06-07 2008-12-11 Astrazeneca Ab Oxadiazole derivatives and their use as metabotropic glutamate receptor potentiators - 842
US7754742B2 (en) 2008-07-18 2010-07-13 Eli Lilly And Company Imidazole carboxamides
WO2010104195A1 (en) * 2009-03-11 2010-09-16 Banyu Pharmaceutical Co.,Ltd. Novel isoindolin-1-one derivative
EP2303872A1 (en) * 2008-06-06 2011-04-06 AstraZeneca AB Isoxazole derivatives and their use as metabotropic glutamate receptor potentiators
US8153638B2 (en) 2005-08-12 2012-04-10 Astrazeneca Ab Metabotropic glutamate-receptor-potentiating isoindolones
US8314120B2 (en) 2010-03-30 2012-11-20 Abbott Gmbh & Co. Kg Small molecule potentiators of metabotropic glutamate receptors
US8664214B2 (en) 2010-03-30 2014-03-04 AbbVie Deutschland GmbH & Co. KG Small molecule potentiators of metabotropic glutamate receptors I
US8691849B2 (en) 2008-09-02 2014-04-08 Janssen Pharmaceuticals, Inc. 3-azabicyclo[3.1.0]hexyl derivatives as modulators of metabotropic glutamate receptors
US8691813B2 (en) 2008-11-28 2014-04-08 Janssen Pharmaceuticals, Inc. Indole and benzoxazine derivatives as modulators of metabotropic glutamate receptors
US8697689B2 (en) 2008-10-16 2014-04-15 Janssen Pharmaceuticals, Inc. Indole and benzomorpholine derivatives as modulators of metabotropic glutamate receptors
US8722894B2 (en) 2007-09-14 2014-05-13 Janssen Pharmaceuticals, Inc. 1,3-disubstituted-4-phenyl-1H-pyridin-2-ones
EP2770997A1 (en) * 2011-10-28 2014-09-03 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor m1
US8841323B2 (en) 2006-03-15 2014-09-23 Janssen Pharmaceuticals, Inc. 1, 4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of MGLUR2-receptors
US8895571B2 (en) 2011-10-14 2014-11-25 Incyte Corporation Isoindolinone and pyrrolopyridinone derivatives as Akt inhibitors
US8906939B2 (en) 2007-03-07 2014-12-09 Janssen Pharmaceuticals, Inc. 3-cyano-4-(4-tetrahydropyran-phenyl)-pyridin-2-one derivatives
US8937060B2 (en) 2009-05-12 2015-01-20 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US8946205B2 (en) 2009-05-12 2015-02-03 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US8993591B2 (en) 2010-11-08 2015-03-31 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a] pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
US9012448B2 (en) 2010-11-08 2015-04-21 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
US9067891B2 (en) 2007-03-07 2015-06-30 Janssen Pharmaceuticals, Inc. 1,4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of mGluR2-receptors
US9085577B2 (en) 2009-05-12 2015-07-21 Janssen Pharmaceuticals, Inc. 7-aryl-1,2,4-triazolo[4,3-A]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US9114138B2 (en) 2007-09-14 2015-08-25 Janssen Pharmaceuticals, Inc. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′] bipyridinyl-2′-ones
US9242933B2 (en) 2007-05-25 2016-01-26 AbbVie Deutschland GmbH & Co. KG Heterocyclic compounds as positive modulators of metabotropic glutamate receptor 2 (mGlu2 receptor)
US9271967B2 (en) 2010-11-08 2016-03-01 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US9708315B2 (en) 2013-09-06 2017-07-18 Janssen Pharmaceutica Nv 1,2,4-triazolo[4,3-a]pyridine compounds and their use as positive allosteric modulators of MGLUR2 receptors
US10106542B2 (en) 2013-06-04 2018-10-23 Janssen Pharmaceutica Nv Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
EP3459939A1 (en) * 2017-09-26 2019-03-27 Pragma Therapeutics Novel heterocyclic compounds as modulators of mglur7
US10537573B2 (en) 2014-01-21 2020-01-21 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US11078204B2 (en) 2018-11-13 2021-08-03 Incyte Corporation Heterocyclic derivatives as PI3K inhibitors
US11161838B2 (en) 2018-11-13 2021-11-02 Incyte Corporation Heterocyclic derivatives as PI3K inhibitors
US11369606B2 (en) 2014-01-21 2022-06-28 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US11396502B2 (en) 2018-11-13 2022-07-26 Incyte Corporation Substituted heterocyclic derivatives as PI3K inhibitors

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080139846A1 (en) * 2006-09-01 2008-06-12 Astrazeneca Ab New Process 298
JP2010525073A (en) * 2007-04-23 2010-07-22 ハウス イアー インスティトゥート Treatment and / or prevention of senile deafness by regulating metabotropic glutamate receptor 7
WO2009004430A1 (en) * 2007-06-29 2009-01-08 Pfizer Inc. N-benzyl oxazolidinones and related heterocycleic compounds as potentiators of glutamate receptors
GB0811643D0 (en) 2008-06-25 2008-07-30 Cancer Rec Tech Ltd New therapeutic agents
RU2557236C2 (en) * 2009-10-09 2015-07-20 Селджин Корпорейшн Method of producing 2-(1-phenylethyl)isoindolin-1-one compounds
WO2011051490A2 (en) 2009-11-02 2011-05-05 N.V. Organon Heterocyclic derivatives
DK2521722T3 (en) * 2010-01-07 2015-07-20 Astrazeneca Ab PROCEDURE FOR THE PREPARATION OF A METABOTROPIC GLUTAMATE RECEPTOR POSITIVE ALLOSTERIC MODULATOR - 874
TWI713455B (en) 2014-06-25 2020-12-21 美商伊凡克特治療公司 Mnk inhibitors and methods related thereto
GB201517217D0 (en) 2015-09-29 2015-11-11 Astex Therapeutics Ltd And Cancer Res Technology Ltd Pharmaceutical compounds
GB201517216D0 (en) 2015-09-29 2015-11-11 Cancer Res Technology Ltd And Astex Therapeutics Ltd Pharmaceutical compounds
EA034440B1 (en) 2015-10-29 2020-02-07 Эффектор Терапьютикс, Инк. PYRROLO-, PYRAZOLO-, IMIDAZO-PYRIMIDINE AND PYRIDINE COMPOUNDS THAT INHIBIT Mnk1 AND Mnk2
AU2016343687A1 (en) 2015-10-29 2018-06-07 Effector Therapeutics, Inc. Isoindoline, azaisoindoline, dihydroindenone and dihydroazaindenone inhibitors of Mnk1 and Mnk2
US10000487B2 (en) 2015-11-20 2018-06-19 Effector Therapeutics, Inc. Heterocyclic compounds that inhibit the kinase activity of Mnk useful for treating various cancers
CN105837557A (en) * 2016-05-05 2016-08-10 青岛辰达生物科技有限公司 Preparation method of alogliptin for treating type-II diabetes
CN106279182B (en) * 2016-07-29 2019-06-07 中国药科大学 A kind of pyrrolo- [2,1-a] iso-indole ketone compounds and its synthetic method
CA3053493A1 (en) 2017-02-14 2018-08-23 Effector Therapeutics, Inc. Piperidine-substituted mnk inhibitors and methods related thereto
GB201704965D0 (en) 2017-03-28 2017-05-10 Astex Therapeutics Ltd Pharmaceutical compounds
GB201704966D0 (en) 2017-03-28 2017-05-10 Astex Therapeutics Ltd Pharmaceutical compounds
CA3117169A1 (en) 2018-10-24 2020-04-30 Effector Therapeutics, Inc. Crystalline forms of mnk inhibitors
CN110498759A (en) * 2019-09-12 2019-11-26 天津瑞岭化工有限公司 The synthetic method of isoindoline ketone compound

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE620654A (en) * 1961-07-28 1963-01-25 May & Baker Ltd New isoindolinones
US3579524A (en) * 1968-06-05 1971-05-18 Miles Lab 2-aminoalkyl derivatives of phthalimidines
US3993617A (en) * 1975-10-30 1976-11-23 Morton-Norwich Products, Inc. Antifungal 2-substituted phthalimidines
EP0548934A1 (en) * 1991-12-25 1993-06-30 Mitsubishi Chemical Corporation Benzamide derivatives
US5681954A (en) * 1993-05-14 1997-10-28 Daiichi Pharmaceutical Co., Ltd. Piperazine derivatives
WO1999026927A2 (en) * 1997-11-21 1999-06-03 Nps Pharmaceuticals, Inc. Metabotropic glutamate receptor antagonists for treating central nervous system diseases
WO2004024702A1 (en) * 2002-08-24 2004-03-25 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel carboxamide compounds having an mch-antagonistic effect, medicaments containing said compounds, and methods for the production thereof
WO2004031178A1 (en) * 2002-10-07 2004-04-15 Pfizer Limited Pyrazole derivatives
WO2005040157A2 (en) * 2003-10-22 2005-05-06 Eli Lilly And Company Novel mch receptor antagonists
WO2005074643A2 (en) * 2004-01-30 2005-08-18 Smithkline Beecham Corporation Benzamide compounds useful as rock inhibitors
WO2005085216A1 (en) * 2004-03-05 2005-09-15 Nissan Chemical Industries, Ltd. Isoxazoline-substituted benzamide compound and noxious organism control agent
WO2005085214A1 (en) * 2004-03-05 2005-09-15 Banyu Pharmaceutical Co., Ltd Diaryl-substituted five-membered heterocycle derivative
WO2006020879A1 (en) * 2004-08-13 2006-02-23 Astrazeneca Ab Isoindolone compounds and their use as metabotropic glutamate receptor potentiators

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3717561A1 (en) * 1987-05-25 1988-12-08 Thomae Gmbh Dr K INDOL, ISOCHINOLINE AND BENZAZEPINE DERIVATIVES, MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS AND METHOD FOR THE PRODUCTION THEREOF
JPH02184667A (en) * 1989-01-11 1990-07-19 Meiji Seika Kaisha Ltd N-n'-di-substituted piperazyl derivative and dysuria-improving agent containing the same
RU2124511C1 (en) * 1993-05-14 1999-01-10 Фармасьютикал Ко., Лтд Piperazine derivatives
IL151533A0 (en) * 2000-02-29 2003-04-10 Mitsubishi Pharma Corp Novel cyclic amide derivatives
DE10031391A1 (en) * 2000-07-03 2002-02-07 Knoll Ag Bicyclic compounds and their use for the prophylaxis and therapy of cerebral ischemia
JP5079202B2 (en) * 2000-07-18 2012-11-21 大日本住友製薬株式会社 Serotonin reuptake inhibitor

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE620654A (en) * 1961-07-28 1963-01-25 May & Baker Ltd New isoindolinones
US3579524A (en) * 1968-06-05 1971-05-18 Miles Lab 2-aminoalkyl derivatives of phthalimidines
US3993617A (en) * 1975-10-30 1976-11-23 Morton-Norwich Products, Inc. Antifungal 2-substituted phthalimidines
EP0548934A1 (en) * 1991-12-25 1993-06-30 Mitsubishi Chemical Corporation Benzamide derivatives
US5681954A (en) * 1993-05-14 1997-10-28 Daiichi Pharmaceutical Co., Ltd. Piperazine derivatives
WO1999026927A2 (en) * 1997-11-21 1999-06-03 Nps Pharmaceuticals, Inc. Metabotropic glutamate receptor antagonists for treating central nervous system diseases
WO2004024702A1 (en) * 2002-08-24 2004-03-25 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel carboxamide compounds having an mch-antagonistic effect, medicaments containing said compounds, and methods for the production thereof
WO2004031178A1 (en) * 2002-10-07 2004-04-15 Pfizer Limited Pyrazole derivatives
WO2005040157A2 (en) * 2003-10-22 2005-05-06 Eli Lilly And Company Novel mch receptor antagonists
WO2005074643A2 (en) * 2004-01-30 2005-08-18 Smithkline Beecham Corporation Benzamide compounds useful as rock inhibitors
WO2005085216A1 (en) * 2004-03-05 2005-09-15 Nissan Chemical Industries, Ltd. Isoxazoline-substituted benzamide compound and noxious organism control agent
WO2005085214A1 (en) * 2004-03-05 2005-09-15 Banyu Pharmaceutical Co., Ltd Diaryl-substituted five-membered heterocycle derivative
WO2006020879A1 (en) * 2004-08-13 2006-02-23 Astrazeneca Ab Isoindolone compounds and their use as metabotropic glutamate receptor potentiators

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
AHN K H ET AL: "N-Substituted-3-arylpyrrolidines: potent and selective ligands at serotonin 1A receptor", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 9, no. 10, 17 May 1999 (1999-05-17), pages 1379 - 1384, XP004164896 *
ANDERSON P S ET AL: "Synthesis of 9,10-dihydroanthracen-9,10-imines", JOURNAL OF ORGANIC CHEMISTRY, vol. 44, no. 9, 1979, pages 1519 - 15, XP002981802 *
BAILEY D M ET AL: "2,3-Diarylphthalimidines", JOURNAL OF MEDICINAL CHEMISTRY, vol. 14, no. 3, 1971, pages 240 - 241, XP002357873 *
BARR N ET AL: "Palladium-assisted organic reactions. VIII. Simple syntheses of 2,3-disubstituted phthalimidines", JOURNAL OF ORGANOMETALLIC CHEMISTRY, vol. 302, no. 1, 11 March 1986 (1986-03-11), pages 117 - 126, XP002383544 *
BÖHME H ET AL: "Untersuchungen in der Phthalimidin-Reihe", DIE PHARMAZIE, no. 25, 1970, pages 283 - 289, XP002357872 *
BREYTENBACH J C ET AL: "Synthesis and antimicrobial activity of some isoindolin-1-ones derivatives", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 10, no. 15, 7 August 2000 (2000-08-07), pages 1629 - 1631, XP004213210 *
CASAGRANDE C ET AL: "Synthesis of some isoindolines and 1,2,3,4-tetrahydroisoquinolines and their evaluation as alpha-adrenergic and adrenergic neuron blocking agents", IL FARMACO, EDIZIONE SCIENTIFICA, vol. 27, no. 6, June 1972 (1972-06-01), pages 445 - 470, XP000571647 *
CLAYDEN J ET AL: "Dearomatizing anionic cyclization of substituted N-cumyl-N-benzyl-benzamides on treatment with LDA: synthesis of partially saturated substituted isoindolones", ORGANIC LETTERS, vol. 2, no. 26, 2000, pages 4229 - 4232, XP002345295 *
COUTURE A ET AL: "Diastereoselective addition of metalated isoindolin-1-ones to aldehydes. stereoselective preparation of (E)-3-arylideneisoindolin-1-ones", TETRAHEDRON LETTERS, vol. 43, no. 12, 18 March 2002 (2002-03-18), pages 2207 - 2210, XP004344002 *
GRIGG R ET AL: "Isoindolinones via a room temperature palladium nanoparticle-catalysed 3-component cyclative carbonylation-amination cascade", TETRAHEDRON LETTERS, vol. 44, no. 37, 8 September 2003 (2003-09-08), pages 6979 - 6982, XP004447066 *
HATT H H ET AL: "Heterocyclic nitrogen compounds. Part II. The preparation of 5:7:12:14-tetrahydro-6:3-diazanaphthacene and some derivatives thereof, with an example of ring expansion and contraction in a Clemmensen reductionth", JOURNAL OF THE CHEMICAL SOCIETY, 1952, pages 199 - 205, XP002357871 *
HOARAU C ET AL: "A versatile synthesis of poly- and diversely substituted isoindolin-1-ones", SYNTHESIS, no. 5, 2000, pages 655 - 660, XP002383545 *
LUZZIO F A ET AL: "A facile scheme for phthalimide - phthalimidine conversion", TETRAHEDRON LETTERS, vol. 40, no. 11, 12 March 1999 (1999-03-12), pages 2087 - 2090, XP002357870 *
MAYER P ET AL: "New substituted 1-(2,3-dihydrobenzo[1,4]dioxin-2-ylmethyl piperidin-4-yl derivatives with alpha2-adrenoreceptor antagonist activity", JOURNAL OF MEDICINAL CHEMISTRY, vol. 43, 19 September 2000 (2000-09-19), pages 3653 - 3664, XP001183987 *
MOREAU A ET AL: "A new approach to isoindoloisoquinolinones. A simple synthesis of nuevamine", TETRAHEDRON, vol. 60, no. 29, 12 July 2004 (2004-07-12), pages 6169 - 6176, XP002357869 *
MORI M ET AL: "Reactions and syntheses with organometallic compounds. 7. Synthesis of benzolactams by palladium-catalyzed amidation", JOURNAL OF ORGANIC CHEMISTRY, vol. 43, no. 9, 28 April 1978 (1978-04-28), pages 1864 - 1867, XP002357874 *
NORMAN M H ET AL: "Conformationally restricted analogues of remoxipride as potential antipsychotic agents", JOURNAL OF MEDICINAL CHEMISTRY, vol. 36, no. 22, 29 October 1993 (1993-10-29), pages 3417 - 3423, XP002357876 *
NORMAN M H ET AL: "Effect of linking bridge modifications on the antipsychotic profile of some phthalimide and isoindolinone derivatives", JOURNAL OF MEDICINAL CHEMISTRY, vol. 39, no. 1, 1996, pages 149 - 157, XP000982309 *
RYS V ET AL: "A short total synthesis of the alkaloids piperolactam C, goniopedaline, and stigmalactam", EUROPEAN JOURNAL OF ORGANIC CHEMISTRY, no. 7, April 2003 (2003-04-01), pages 1231 - 1237, XP002383546 *
SUGIMOTO H ET AL: "Synthesis and structure-activity relationships of acetylcholinesterase inhibitors: 1-benzyl-4-(2-phthalimidoethyl)piperidine and related derivatives", JOURNAL OF MEDICINAL CHEMISTRY, vol. 35, no. 24, 1992, pages 4542 - 4548, XP002319779 *
YAMAMOTO Y ET AL: "Synthesis of benzo-fused lactams and lactones via Ru(II)-catalyzed cycloaddition of amide- and ester-tethered alpha,omega-diyines with terminal alkynes: electronic directing effect of internal conjugated carbonyl groups", ORGANIC AND BIOMOLECULAE CHEMISTRY, vol. 2, no. 9, May 2004 (2004-05-01), pages 1287 - 1294, XP002357868 *
ZHUANG Z-P ET AL: "Isoindol-1-one analogues of 4-(2'-methoxyphenyl)1- [2'-[N-(2''-pyridyl)-p-iodobenzamido] ethyl]piperazine (p-MPPI) as 5-HT1A receptor ligands", JOURNAL OF MEDICINAL CHEMISTRY, vol. 41, no. 2, 15 January 1998 (1998-01-15), pages 157 - 166, XP002357875 *

Cited By (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8153638B2 (en) 2005-08-12 2012-04-10 Astrazeneca Ab Metabotropic glutamate-receptor-potentiating isoindolones
US9266834B2 (en) 2006-03-15 2016-02-23 Janssen Pharmaceuticals, Inc. 1, 4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of MGLUR2-receptors
US8841323B2 (en) 2006-03-15 2014-09-23 Janssen Pharmaceuticals, Inc. 1, 4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of MGLUR2-receptors
US8906939B2 (en) 2007-03-07 2014-12-09 Janssen Pharmaceuticals, Inc. 3-cyano-4-(4-tetrahydropyran-phenyl)-pyridin-2-one derivatives
US9067891B2 (en) 2007-03-07 2015-06-30 Janssen Pharmaceuticals, Inc. 1,4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of mGluR2-receptors
WO2008130853A1 (en) * 2007-04-17 2008-10-30 Astrazeneca Ab Hydrazides and their use as metabotropic glutamate receptor potentiators - 681
US9242933B2 (en) 2007-05-25 2016-01-26 AbbVie Deutschland GmbH & Co. KG Heterocyclic compounds as positive modulators of metabotropic glutamate receptor 2 (mGlu2 receptor)
WO2008150233A1 (en) * 2007-06-07 2008-12-11 Astrazeneca Ab Oxadiazole derivatives and their use as metabotropic glutamate receptor potentiators - 842
US7799792B2 (en) 2007-06-07 2010-09-21 Astrazeneca Ab Metabotropic glutamate receptor oxadiazole ligands and their use as potentiators 841
EP2444399A1 (en) * 2007-06-07 2012-04-25 AstraZeneca AB Oxadiazole derivatives and their use as metabotropic glutamate receptor potentiators - 842
WO2008150232A1 (en) * 2007-06-07 2008-12-11 Astrazeneca Ab Metabotropic glutamate receptor oxadiazole ligands and their use as potentiators - 841
RU2470931C2 (en) * 2007-06-07 2012-12-27 Астразенека Аб Oxadiazole derivatives and use thereof as metabotropic glutamate receptor potentiators 842
US8377939B2 (en) 2007-06-07 2013-02-19 Astrazeneca Ab Oxadiazole derivatives and their use as metabotropic glutamate receptor potentiators 842
US8377940B2 (en) 2007-06-07 2013-02-19 Astrazeneca Ab Oxadiazole derivatives and their use as metabotropic glutamate receptor potentiators—842
US9132122B2 (en) 2007-09-14 2015-09-15 Janssen Pharmaceuticals, Inc. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′]bipyridinyl-2′-ones
US9114138B2 (en) 2007-09-14 2015-08-25 Janssen Pharmaceuticals, Inc. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′] bipyridinyl-2′-ones
US8722894B2 (en) 2007-09-14 2014-05-13 Janssen Pharmaceuticals, Inc. 1,3-disubstituted-4-phenyl-1H-pyridin-2-ones
US11071729B2 (en) 2007-09-14 2021-07-27 Addex Pharmaceuticals S.A. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′]bipyridinyl-2′-ones
US8148372B2 (en) 2008-06-06 2012-04-03 Astrazeneca Ab Metabotropic glutamate receptor isoxazole ligands and their use as potentiators—286
TWI477499B (en) * 2008-06-06 2015-03-21 Astrazeneca Ab Metabotropic glutamate receptor isoxazole ligands and their use as potentiators-286
EP2772492A1 (en) * 2008-06-06 2014-09-03 AstraZeneca AB Use of an isoxazole derivative for the treatment of withdrawal symptom
EP2303872A1 (en) * 2008-06-06 2011-04-06 AstraZeneca AB Isoxazole derivatives and their use as metabotropic glutamate receptor potentiators
CN102105466A (en) * 2008-06-06 2011-06-22 阿斯利康(瑞典)有限公司 Isoxazole derivatives and their use as metabotropic glutamate receptor potentiators
EP2303872A4 (en) * 2008-06-06 2012-03-21 Astrazeneca Ab Isoxazole derivatives and their use as metabotropic glutamate receptor potentiators
US7754742B2 (en) 2008-07-18 2010-07-13 Eli Lilly And Company Imidazole carboxamides
US8691849B2 (en) 2008-09-02 2014-04-08 Janssen Pharmaceuticals, Inc. 3-azabicyclo[3.1.0]hexyl derivatives as modulators of metabotropic glutamate receptors
US8697689B2 (en) 2008-10-16 2014-04-15 Janssen Pharmaceuticals, Inc. Indole and benzomorpholine derivatives as modulators of metabotropic glutamate receptors
US8691813B2 (en) 2008-11-28 2014-04-08 Janssen Pharmaceuticals, Inc. Indole and benzoxazine derivatives as modulators of metabotropic glutamate receptors
US8362052B2 (en) 2009-03-11 2013-01-29 Msd K.K. Isoindolin-1-one derivative
JP2012520240A (en) * 2009-03-11 2012-09-06 Msd株式会社 Novel isoindoline-1-one derivatives
WO2010104195A1 (en) * 2009-03-11 2010-09-16 Banyu Pharmaceutical Co.,Ltd. Novel isoindolin-1-one derivative
US8946205B2 (en) 2009-05-12 2015-02-03 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US8937060B2 (en) 2009-05-12 2015-01-20 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US9737533B2 (en) 2009-05-12 2017-08-22 Janssen Pharmaceuticals. Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US10071095B2 (en) 2009-05-12 2018-09-11 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of neurological and psychiatric disorders
US9085577B2 (en) 2009-05-12 2015-07-21 Janssen Pharmaceuticals, Inc. 7-aryl-1,2,4-triazolo[4,3-A]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US9226930B2 (en) 2009-05-12 2016-01-05 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-a] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US8314120B2 (en) 2010-03-30 2012-11-20 Abbott Gmbh & Co. Kg Small molecule potentiators of metabotropic glutamate receptors
US8664214B2 (en) 2010-03-30 2014-03-04 AbbVie Deutschland GmbH & Co. KG Small molecule potentiators of metabotropic glutamate receptors I
US9012448B2 (en) 2010-11-08 2015-04-21 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
US8993591B2 (en) 2010-11-08 2015-03-31 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a] pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
US9271967B2 (en) 2010-11-08 2016-03-01 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US8895571B2 (en) 2011-10-14 2014-11-25 Incyte Corporation Isoindolinone and pyrrolopyridinone derivatives as Akt inhibitors
EP2770997A1 (en) * 2011-10-28 2014-09-03 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor m1
EP3639823A1 (en) * 2011-10-28 2020-04-22 Vanderbilt University Center for Technology Transfer and Commercialization Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor m1
EP2770997A4 (en) * 2011-10-28 2015-04-01 Univ Vanderbilt Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor m1
AU2012328476B2 (en) * 2011-10-28 2017-03-30 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor M1
US9586964B2 (en) 2011-10-28 2017-03-07 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor M1
AU2019200952B2 (en) * 2011-10-28 2020-07-16 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor M1
AU2017204409B2 (en) * 2011-10-28 2018-11-15 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor M1
US10654847B2 (en) 2011-10-28 2020-05-19 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor M1
EP3153167A1 (en) * 2011-10-28 2017-04-12 Vanderbilt University Substituted 2-(4-heterocyclylbenzyl)isoindolin-1-one analogs as positive allosteric modulators of the muscarinic acetylcholine receptor m1
US10584129B2 (en) 2013-06-04 2020-03-10 Janssen Pharmaceuticals Nv Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
US10106542B2 (en) 2013-06-04 2018-10-23 Janssen Pharmaceutica Nv Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
US9708315B2 (en) 2013-09-06 2017-07-18 Janssen Pharmaceutica Nv 1,2,4-triazolo[4,3-a]pyridine compounds and their use as positive allosteric modulators of MGLUR2 receptors
US11369606B2 (en) 2014-01-21 2022-06-28 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US10537573B2 (en) 2014-01-21 2020-01-21 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US11103506B2 (en) 2014-01-21 2021-08-31 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
CN111148744A (en) * 2017-09-26 2020-05-12 布拉格玛治疗公司 Novel heterocyclic compounds as MGLUR7 modulators
EP3459939A1 (en) * 2017-09-26 2019-03-27 Pragma Therapeutics Novel heterocyclic compounds as modulators of mglur7
WO2019063596A1 (en) * 2017-09-26 2019-04-04 Pragma Therapeutics Novel heterocyclic compounds as modulators of mglur7
US11414395B2 (en) 2017-09-26 2022-08-16 Pragma Therapeutics Heterocyclic compounds as modulators of mGluR7
US11078204B2 (en) 2018-11-13 2021-08-03 Incyte Corporation Heterocyclic derivatives as PI3K inhibitors
US11161838B2 (en) 2018-11-13 2021-11-02 Incyte Corporation Heterocyclic derivatives as PI3K inhibitors
US11396502B2 (en) 2018-11-13 2022-07-26 Incyte Corporation Substituted heterocyclic derivatives as PI3K inhibitors

Also Published As

Publication number Publication date
JP2009509921A (en) 2009-03-12
EP1912939A1 (en) 2008-04-23
CN101277934A (en) 2008-10-01
EP1912940A1 (en) 2008-04-23
JP5031745B2 (en) 2012-09-26
WO2007021309A1 (en) 2007-02-22
CN101309905A (en) 2008-11-19
JP2009509920A (en) 2009-03-12

Similar Documents

Publication Publication Date Title
WO2007021308A1 (en) Metabotropic glutamate-receptor-potentiating isoindolones
US7868008B2 (en) Substituted isoindolones and their use as metabotropic glutamate receptor potentiators
US8153638B2 (en) Metabotropic glutamate-receptor-potentiating isoindolones
TWI417100B (en) Oxadiazole derivatives and their use as metabotropic glutamate receptor potentiators-842
US20110053953A1 (en) AZA-Isoindolones and Their Use as Metabotropic Glutamate Receptor Potentiators - 613
WO2008130853A1 (en) Hydrazides and their use as metabotropic glutamate receptor potentiators - 681
JP5031565B2 (en) Isoindole compounds and their use as metabotropic glutamate receptor potentiators
JP5608655B2 (en) Modulator of P2X3 receptor activity
DE69929704T2 (en) TETRAHYDROBENZAZEPINE DERIVATIVES USE AS DOPAMINE D3 RECEPTOR MODULATORS (ANTIPSYCHOTIC AGENTS)
WO2008032191A2 (en) Spiro-oxazolidinone compounds and their use as metabotropic glutamate receptor potentiators
JP2009519929A (en) Oxazolidinone compounds and their use as metabotropic glutamate receptor potentiators
JP2010520876A (en) Piperazine and piperidine mGluR5 potentiator
JP2009503069A (en) Tricyclic benzimidazoles and their use as modulators of metabolic glutamate receptors
JP2009532381A (en) Bicyclic benzimidazole compounds and use of the compounds as metabotropic glutamate receptor potentiators
MX2010012357A (en) Amide compound.
JP2010529117A (en) Metabotropic glutamate receptor oxadiazole ligands and their use as potentiators
JP2006523707A (en) Therapeutic compound
JP2009536213A (en) Fused heterocyclic compounds and their use as MGLUR5 modulators
DE60305332T2 (en) IMIDAZOi1,2-AöPYRIDINE

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680036311.7

Country of ref document: CN

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

Ref document number: 934/DELNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008525976

Country of ref document: JP

Ref document number: 2006720758

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12063007

Country of ref document: US