WO2007018998A2 - Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators - Google Patents

Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators Download PDF

Info

Publication number
WO2007018998A2
WO2007018998A2 PCT/US2006/028165 US2006028165W WO2007018998A2 WO 2007018998 A2 WO2007018998 A2 WO 2007018998A2 US 2006028165 W US2006028165 W US 2006028165W WO 2007018998 A2 WO2007018998 A2 WO 2007018998A2
Authority
WO
WIPO (PCT)
Prior art keywords
dihydro
methyl
imidazo
fluoro
quinoline
Prior art date
Application number
PCT/US2006/028165
Other languages
French (fr)
Other versions
WO2007018998A3 (en
Inventor
Methvin Isaac
Abdelmalik Slassi
Ian Egle
Fupeng Ma
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
Application filed by Astrazeneca Ab filed Critical Astrazeneca Ab
Priority to EP06787957A priority Critical patent/EP1912989A2/en
Priority to US11/996,727 priority patent/US20080318999A1/en
Priority to CA002616020A priority patent/CA2616020A1/en
Priority to BRPI0614168A priority patent/BRPI0614168A2/en
Priority to AU2006279034A priority patent/AU2006279034A1/en
Priority to JP2008524995A priority patent/JP2009503069A/en
Priority to MX2008001152A priority patent/MX2008001152A/en
Publication of WO2007018998A2 publication Critical patent/WO2007018998A2/en
Publication of WO2007018998A3 publication Critical patent/WO2007018998A3/en
Priority to IL188809A priority patent/IL188809A0/en
Priority to NO20080475A priority patent/NO20080475L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/06Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/06Peri-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • 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
    • A61P25/08Antiepileptics; Anticonvulsants
    • A61P25/10Antiepileptics; Anticonvulsants for petit-mal
    • 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
    • A61P25/28Drugs 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • 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
    • A61P9/10Drugs 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/06Peri-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/582Recycling of unreacted starting or intermediate materials

Definitions

  • the present invention relates to novel compounds that function as modulators of glutamate receptors, methods for their preparation, pharmaceutical compositions containing them and their use in therapy.
  • the metabotropic glutamate receptors constitute a family of GTP-binding-protein (G-protein) coupled receptors that are activated by glutamate, and have important roles in synaptic activity in the central nervous system, including 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 ah, 1993, Trends Pharmacol.
  • PI phosphoinositide
  • Group-I includes niGluRl and mGluR5, which activate phospholipase C and the generation of an intracellular calcium signal.
  • the Group-II mGluR2 and mGluR3
  • Group-Ill mGluR4, mGluR ⁇ , niGluR7, and mGluR8 mGluRs mediate an inhibition of adenylyl cyclase activity and cyclic AMP levels.
  • mGluRs Members of the mGluR family of receptors are 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 al, 1994, Nature, 368:740 ; Aiba et al, 1994, Cell, 79:365 ; Aiba et al, 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 al, 1995, Neuropharmacology, supra; Knopfel et al, 1995, J. Med. Chem., 38:1417).
  • One embodiment of the invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof:
  • A is selected from the group consisting Of CR 8 R 9 , NR 5 , O, S, SO and SO 2 ;
  • B is selected from the group consisting of CH and N;
  • D is selected from the group consisting of NH, N-Ci -6 -alkyl, and -(CR 5 R 6 ) Z -, wherein one of the -CR 5 R 6 - groups maybe replaced by -C(O)-, NH, orNC 1-6 -alkyl;
  • L is selected from the group consisting of a direct bond and -(CR 5 R 6 ) W -, wherein when L is -(CR 5 R 6 V:
  • B-L may be unsaturated, or two adjacent carbon atoms may form part of a cyclopropyl ring;
  • one or two CR 5 R 6 groups may be replaced with O, S, or NR 5 ;
  • ring represents a ring selected from the group consisting of azetidine and a 5- to 7- membered ring, which may be unsaturated, wherein the ring may be substituted by one or more R 4 ;
  • R 1 in each instance, is selected from the group consisting of H, F, Cl, Br, I, OH, CN, nitro, C 1-6 -alkyl, OC 1-6 -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 3-8 -cycloalkyl, C 1-6 -alkylene-C 3-8 -cycloalkyl, OCo -6 -alkylene-C 3-8 -cycloalkyl, aryl, heteroaryl, C 1-6 -alkylenearyl, C 1-6 - alkyleneheteroaryl, OC 1-6 -alkylenearyl, OC 1-6 -alkyleneheteroaryl, C 1-6 - alkyleneheterocycloalkyl, (CO)R
  • R 2 and R 4 are independently selected from the group consisting of H, F, Cl, Br, L CN, nitro, hydroxy, oxo, Ci -6 -alkyl, OCi -6 -alkyl, C 1-6 -alkylhalo, OC 1-6 - alkylhalo, and C 0-6 -alkyleneNR 5 R 6 ;
  • R 3 is a 5- to 12-membered ring system that is optionally substituted by up to three R 1 groups, wherein the ring system may contain one or more heteroatoms independently selected from the group consisting of N, O and S;
  • R 5 is selected from the group consisting of H, C 1-6 -alkyl, aryl, C 3-8 -cycloalkyl, Q -6 - alkylenearyl and Q-e-alkylene-Cs-g-cycloalkyl, wherein any cyclic group may be further substituted with one or more independently-selected R 2 groups;
  • R 6 and R 7 are independently selected from the group consisting of H and C 1-6 -alkyl
  • R 8 and R 9 are independently selected from the group consisting of H, -O-(CH 2 ) 2 -O- and - O-(CH 2 ) 3 -O-;
  • n and n are integers independently selected from the group consisting of 0, 1, 2, 3 and 4, with the proviso that m and n cannot simultaneously be 0;
  • x andy are integers independently selected from the group consisting of 1, 2, and 3; and w and z are integers independently selected from the group consisting of 1, 2, 3, 4, 5, and 6; or a pharmaceutically-acceptable salt, hydrate, solvate, isoform, tautomer, optical isomer, or combination thereof.
  • the invention also provides, in addition to a compound of formula I, a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
  • Another embodiment of the invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to formula I together with a pharmaceutically acceptable carrier or excipient.
  • Yet another embodiment of the invention is a method for treating or preventing a neurological and psychiatric disorder that is associated with glutamate dysfunction.
  • the method comprises the step of administering, to a subject in need of the treatment, a therapeutically effective amount of a compound of formula I, typically in the form of a pharmaceutical composition thereof.
  • Still another embodiment of the invention is the use of a compound 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 herein.
  • Another embodiment of the invention provides a compound of formula I, or a pharmaceutically acceptable salt or solvate thereof, for use in therapy.
  • the invention additionally provides a process for the preparation of compounds of formula I. General and specific processes are discussed in more detail below.
  • the present invention relates to the discovery of compounds that exhibit activity as pharmaceuticals, in particular as modulators of metabotropic glutamate receptors. More particularly, the compounds of the present invention exhibit activity as potentiators, more particularly positive allosteric modulators, of the mGluR2 receptor, and are useful in therapy, in particular for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction.
  • C 1-6 alkyl as used herein means a straight- or branched-chain hydrocarbon radical having from one to six carbon atoms, and includes methyl, ethyl, propyl, isopropyl, t-butyl and the like.
  • C 2-6 alkenyl as used herein means a straight- or branched-chain alkenyl radical having from two to six carbon atoms, and includes ethenyl, 1-propenyl, 1-butenyl and the like.
  • C 2-6 alkynyl as used herein means a straight- or branched-chain alkynyl radical having from two to six carbon atoms, and includes 1-propynyl (propargyl), 1-butynyl and the like.
  • C 3-8 cycloalkyl as used herein means a cyclic group (which may be unsaturated) having from three to eight carbon atoms, and includes cyclopropyl, cyclohexyl, cyclohexenyl and the like.
  • heterocycloalkyl as used herein means a three- to eight-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 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.
  • alkylene as used herein means a difunctional branched or unbranched saturated hydrocarbon radical having one to six carbon atoms, and includes methylene, ethylene, n- propylene, n-butylene and the like.
  • alkenylene as used herein means a difunctional branched or unbranched hydrocarbon radical having two to six carbon atoms and having at least one double bond, and includes ethenylene, n-propenylene, n-butenylene and the like.
  • alkynylene as used herein means a difunctional branched or unbranched hydrocarbon radical having two to six carbon atoms and having at least one triple bond, and includes ethynylene, n-propynylene, n-butynylene and the like.
  • aryl as used herein means an aromatic group having 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.
  • 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 12-membered ring system ... wherein the ring system may contain one or more heteroatoms independently selected from N, O or S” includes aromatic and heteroaromatic rings, as well as carbocyclic and heterocyclic rings which may be saturated or unsaturated, and which may be mono-, bi- or tri-cyclic, and includes furyl, isoxazolyl, oxazolyl, pyridyl, pyrimidyl, pyrrolyl, thiazolyl, thienyl, triazolyl, morpholinyl, piperazinyl, piperidyl, tetrahydropyranyl, phenyl, cyclohexyl, cyclopentyl, cyclohexanyl, naphthyl, quinolinyl, indolyl, norbornyl, azabicyclooctyl, adamantyl 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.
  • therapeutically 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 , R , R , A, D, B, m, n, x, and y are defined as described above.
  • variable m is 0 and variable n is 2.
  • A is selected from the group consisting of CH 2 and O.
  • D is -(CR 5 R 6 ) Z -.
  • z is preferably 1.
  • each of R 5 and R 6 is H.
  • e presents a piperidine ring.
  • R 3 being a 5- to 7-membered ring that is optionally substituted by 1-3 R 1 groups.
  • the ring may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
  • R is phenyl that is optionally substituted by 1-3 R 1 .
  • R 1 is selected from the group consisting of H, F, Cl, Br, I, nitro, Ci -6 -alkyl, C 1-6 -alkylhalo, C 1-6 -alkylhalo, OQ- 6 -alkylhalo, aryl, C 1-6 -alkylenearyl, and OCi -6 -alkylenearyl, while R is selected from H and Ci -6 -alkyl.
  • compounds of the present invention When compounds of the present invention contain one or more chiral centers, those compounds 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.
  • 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.
  • the present invention encompasses tautomers of the compounds of formula I.
  • salts of the compounds of formula I 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 sufficiently basic compound for example an alkyl amine
  • a suitable acid for example, HCl or acetic acid
  • alkali metal such as sodium, potassium, or lithium
  • 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, in particular an acid addition salt such as a hydrochloride, hydrobromide, phosphate, acetate, fumarate, maleate, tartrate, citrate, methanesulphonate orjy-toluenesulphonate.
  • an acid addition salt such as a hydrochloride, hydrobromide, phosphate, acetate, fumarate, maleate, tartrate, citrate, methanesulphonate orjy-toluenesulphonate.
  • LG in Scheme 1 above represents a leaving group that is capable of being displaced by precursor (ii). Suitable leaving groups are well known in the art and thus include but are not limited to chloride, bromide, and sulphate esters such as mesylate and tosylate.
  • Precursor (i) can be prepared by a number of processes as described in more detail below. An exemplary process may be selected and/or adapted according to the particular structural features of a given precursor (i).
  • Scheme 2 illustrates one exemplary process for making precursor (i).
  • 6-Fluoro-2-methyl-l,2,3,4-tetrahydroquinoline was formylated using the mixed formic/acetic anhydride.
  • This amide was then regioselectively nitrated with nitronium tetrafluoroborate.
  • the formyl group was removed under basic hydrolytic conditions, and the nitro group was reduced to the aniline using hydrogen gas and palladium on carbon.
  • the benzimidazole ring system was finally formed using chloroacetic acid or an equivalent in the presence of a mineral acid catalyst.
  • Scheme 3 illustrates another method for synthesizing precursor (i).
  • 4-fluoroaniline was N- protected with a Boc-protecting group. This group was then used to direct ortho-lithiation, and subsequent trapping with 3-chloro-l-iodopropane provided 6-fluoro-3,4-dihydro-2H- quinoline-1-carboxylic acid tert-butyl ester.
  • the Boc group was replaced with a formyl group, and the synthesis proceeded in an analogous fashion to Scheme 2.
  • Scheme 4 illustrates another method for synthesizing precursor (i). Amino-3-nitrophenol was reacted with 1,2-dibromoethane under basic conditions to produce 5-nitro-3,4-dihydro-2H- 1,4-benzoxazine. The synthesis then proceeded as outlined in Scheme 2.
  • Scheme 5 illustrates another method for synthesizing precursor (i). 8-Nitroquinolone was reduced using hydrogen gas and platinum oxide catalyst. The resultant product was cyclized to the benzimidazole using chloroacetic acid or an equivalent in the presence of a mineral acid catalyst.
  • Scheme 6 illustrates another method for synthesizing precursor (i). This scheme is analogous to Scheme 2.
  • Precursor (ii) in Scheme 1 can be obtained from commercial sources or otherwise synthesized by using well known synthetic methodologies.
  • precursor (ii) can be prepared, if necessary, by the route depicted in Scheme 7 below.
  • Boc-protected 4-piperidone was transformed to the vinyl triflate, which could in turn be converted into the cyclic boronate ester using standard conditions.
  • This intermediate underwent Suzuki reaction conditions with various aryl halides in the presence of a palladium catalyst, yielding the final compounds after deprotection.
  • 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 association 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.
  • 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.
  • 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. Medical Use
  • 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, more particularly as positive allosteric modulators, of the mGluR2 receptor, and are useful in therapy, in particular for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction in an animal.
  • the neurological and psychiatric disorders 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 Chorea, 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
  • 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.
  • a compound 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, intrathoracically, 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 standardization 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.
  • 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 ah, 1992, Neuron, 8:169; Miller et ah, 1995, J. Neuroscience, 15:6103; Balazs, et ah, 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+ ]; 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 min. 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 (O.Ol ⁇ 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 niGluR2 receptor activation.
  • the allosteric activator activity of compounds at the human mGluR2 receptor was 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 [ 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 (3nM to 300 ⁇ M) for 15 min. at room temperature prior to the addition of 1 ⁇ M glutamate, and incubated for 30 min at 3O 0 C in 500 ⁇ assay buffer (20 mM HEPES, 10OmM NaCl, 1OmM MgCl 2 ), containing 30 ⁇ M GDP and O.lnM [ 35 S]-GTPyS (1250 Ci/mmol). Reactions were carried out in triplicate in 2 ml polypropylene 96-well plates.
  • PCC pyridinium chlorochromate ppm parts per million
  • the compounds of the present invention were active in the assays described herein at concentrations (or with ECs 0 values) of less than 10 ⁇ M.
  • Preferred compounds of the invention have EC 50 values of less than 1 ⁇ M; more preferred compounds of less than about 100 nJVL
  • the compounds of Examples 26.55, 26.56, 26.65, 26.69, and 28.1 have EC 50 values of 0.37, 1.58, 0.08, 0.23, and 1.11 ⁇ M, respectively.
  • Lithium aluminum hydride (1.07g, 22.4mmol) was added to the suspension of 7-chloro-5- nitro-4H-benzo[l,4]oxazin-3-one (1.2g, 5.3mmol) in THF (30 mL). The reaction mixture was stirred at room temperature for overnight. The reaction mixture was then quenched with water, the aqueous phase was extracted with ethyl acetate; combined organic phases were washed with water and brine, dried over anhydrous sodium sulfate and concentrated in vacuo.
  • Example 29.1 4-Fluoro-l-[4-(4-fluoro-phenyl)-piperidm-l-ylmethyl]-8,9-dihydro-7H-2,7,9a- triaza-benzo[cd]azulen-6-one
  • Example 30.1 (i) [4R]- and (U) [4S]-8-Fluoro-4-methyl-2- ⁇ [4(4-trifluoromethyl- phenyl)piperizin-l-yl]niethyl ⁇ -5,6-dihydro-4H-imidazo[4,5,l-ij]quinoline
  • Racemic 8-fluoro-4-niethyl-2- ⁇ [4(4-trifluoromethyl-phenyl)piperizin-l-yl]metliyl ⁇ -5,6- dihydro-4H-imidazo[4,5,l-ij]quinoline was separated into its constituent enantiomers using HPLC on a 21.4 mm x 250 mm Chiralcel OJ column. Elution was effected with 25% EtOH in petroleum ether at 15 mL/min.

Landscapes

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

Abstract

The invention provides for a compound of formula (I) or a pharmaceutically acceptable salt thereof; wherein R1, R2, R3, A, B, D, m, n, x, and y are defined as described in the specification. The invention additionally provides a pharmaceutically composition comprising the compound of formula (I), together with a method of using the same to treat or prevent neurological and psychiatric disorders. The compounds are useful in therapy related to the treatment or prevention of mGluR2 receptor-mediated disorders.

Description

TRICYCLIC BENZIMID AZOLES AND THEIR USE AS METABOTROPIC GLUTAMATE RECEPTOR MODULATORS
BACKGROUND OF THE INVENTION
The present invention relates to novel compounds that function as modulators of glutamate receptors, methods for their preparation, pharmaceutical compositions containing them and their use in therapy.
The metabotropic glutamate receptors (mGluR) constitute a family of GTP-binding-protein (G-protein) coupled receptors that are activated by glutamate, and have important roles in synaptic activity in the central nervous system, including 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 ah, 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, which are divided into three groups based upon ^primary sequence similarity, signal transduction linkages, and pharmacological profile. Group-I includes niGluRl and mGluR5, which activate phospholipase C and the generation of an intracellular calcium signal. The Group-II (mGluR2 and mGluR3) and Group-Ill (mGluR4, mGluRό, niGluR7, 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.
Members of the mGluR family of receptors are 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 al, 1994, Nature, 368:740 ; Aiba et al, 1994, Cell, 79:365 ; Aiba et al, 1994, Cell, 79:377). A role for mGluR activation in nociception and analgesia also has been demonstrated (Meller et al, 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 al, 1995, Neuropharmacology, supra; 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
One embodiment of the invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0001
(I) wherein
A is selected from the group consisting Of CR8R9, NR5, O, S, SO and SO2;
B is selected from the group consisting of CH and N; D is selected from the group consisting of NH, N-Ci-6-alkyl, and -(CR5R6)Z-, wherein one of the -CR5R6- groups maybe replaced by -C(O)-, NH, orNC1-6-alkyl;
L is selected from the group consisting of a direct bond and -(CR5R6)W-, wherein when L is -(CR5R6V:
(i) B-L may be unsaturated, or two adjacent carbon atoms may form part of a cyclopropyl ring; or
(ii) one or two CR5R6 groups may be replaced with O, S, or NR5;
Figure imgf000004_0001
represents a ring selected from the group consisting of azetidine and a 5- to 7- membered ring, which may be unsaturated, wherein the ring may be substituted by one or more R4;
R1, in each instance, is selected from the group consisting of H, F, Cl, Br, I, OH, CN, nitro, C1-6-alkyl, OC1-6-alkyl, C1-6-alkylhalo, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6- alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, C1-6-alkylene-C3-8-cycloalkyl, OCo-6-alkylene-C3-8-cycloalkyl, aryl, heteroaryl, C1-6-alkylenearyl, C1-6- alkyleneheteroaryl, OC1-6-alkylenearyl, OC1-6-alkyleneheteroaryl, C1-6- alkyleneheterocycloalkyl, (CO)R5, (CO)OR5, C1-6-alkylene0R5, OC2-6-alkyleneOR5, C1-6-alkylene(CO)R5, OC1-6-alkylene(CO)R5, C1-6-alkylenecyano, OC2-6- alkylenecyano, C0-6-alkyleneNR6R7, OC2-6-alkyleneNR6R7, C1-6-alkylene(CO)NR6R7, OC1-6-alkylene(CO)NR6R7, C0-6-alkyleneNR6(CO)R7, OC2-6-alkyleneNR6(CO)R7, C0- 6-allcyleneNR6(CO)NR6R7, C0-6-alkyleneSO2R5, OC2-6-alkyleneSO2R5, C0-6- alkylene(SO2)NR6R7, OC2-6-alkylene(SO2)NR6R7, C0-6-alkyleneNR6(SO2)R7, OC2-6- alkyleneNR6(S02)R7, Co-6-alkyleneNR6(S02)NR6R7, OC2-6-alkyleneNR6(S02)NR6R7, (CO)NR6R7and SO3R5, wherein any cyclic group may be further substituted with one or more R2;
R2 and R4, in each instance, are independently selected from the group consisting of H, F, Cl, Br, L CN, nitro, hydroxy, oxo, Ci-6-alkyl, OCi-6-alkyl, C1-6-alkylhalo, OC1-6- alkylhalo, and C0-6-alkyleneNR5R6; R3 is a 5- to 12-membered ring system that is optionally substituted by up to three R1 groups, wherein the ring system may contain one or more heteroatoms independently selected from the group consisting of N, O and S;
R5 is selected from the group consisting of H, C1-6-alkyl, aryl, C3-8-cycloalkyl, Q-6- alkylenearyl and Q-e-alkylene-Cs-g-cycloalkyl, wherein any cyclic group may be further substituted with one or more independently-selected R2 groups;
R6 and R7 are independently selected from the group consisting of H and C1-6-alkyl;
R8 and R9 are independently selected from the group consisting of H, -O-(CH2)2-O- and - O-(CH2)3-O-;
m and n are integers independently selected from the group consisting of 0, 1, 2, 3 and 4, with the proviso that m and n cannot simultaneously be 0;
x andy are integers independently selected from the group consisting of 1, 2, and 3; and w and z are integers independently selected from the group consisting of 1, 2, 3, 4, 5, and 6; or a pharmaceutically-acceptable salt, hydrate, solvate, isoform, tautomer, optical isomer, or combination thereof.
The invention also provides, in addition to a compound of formula I, a pharmaceutically acceptable salt, hydrate, solvate, optical isomer, or combination thereof.
Another embodiment of the invention is to provide a pharmaceutical composition comprising a compound according to formula I together with a pharmaceutically acceptable carrier or excipient.
Yet another embodiment of the invention is a method for treating or preventing a neurological and psychiatric disorder that is associated with glutamate dysfunction. The method comprises the step of administering, to a subject in need of the treatment, a therapeutically effective amount of a compound of formula I, typically in the form of a pharmaceutical composition thereof.
Still another embodiment of the invention is the use of a compound 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 herein.
Another embodiment of the invention provides a compound of formula I, or a pharmaceutically acceptable salt or solvate thereof, for use in therapy.
The invention additionally provides a process for the preparation of compounds of formula I. General and specific processes are discussed in more detail below.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention relates to the discovery of compounds that exhibit activity as pharmaceuticals, in particular as modulators of metabotropic glutamate receptors. More particularly, the compounds of the present invention exhibit activity as potentiators, more particularly positive allosteric modulators, of the mGluR2 receptor, and are useful in therapy, in particular for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction.
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 (Pergamon Press, 1979), Sections A, B, C, D, E, F, and H. 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 "C1-6alkyl" as used herein means a straight- or branched-chain hydrocarbon radical having from one to six carbon atoms, and includes methyl, ethyl, propyl, isopropyl, t-butyl and the like. The term "C2-6alkenyl" as used herein means a straight- or branched-chain alkenyl radical having from two to six carbon atoms, and includes ethenyl, 1-propenyl, 1-butenyl and the like.
The term "C2-6alkynyl" as used herein means a straight- or branched-chain alkynyl radical having from two to six carbon atoms, and includes 1-propynyl (propargyl), 1-butynyl and the like.
The term "C3-8cycloalkyl" as used herein means a cyclic group (which may be unsaturated) having from three to eight carbon atoms, and includes cyclopropyl, cyclohexyl, cyclohexenyl and the like.
The term "heterocycloalkyl" as used herein means a three- to eight-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 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 "alkylene" as used herein means a difunctional branched or unbranched saturated hydrocarbon radical having one to six carbon atoms, and includes methylene, ethylene, n- propylene, n-butylene and the like.
The term "alkenylene" as used herein means a difunctional branched or unbranched hydrocarbon radical having two to six carbon atoms and having at least one double bond, and includes ethenylene, n-propenylene, n-butenylene and the like.
The term "alkynylene" as used herein means a difunctional branched or unbranched hydrocarbon radical having two to six carbon atoms and having at least one triple bond, and includes ethynylene, n-propynylene, n-butynylene and the like.
The term "aryl" as used herein means an aromatic group having 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 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 12-membered ring system ... wherein the ring system may contain one or more heteroatoms independently selected from N, O or S" includes aromatic and heteroaromatic rings, as well as carbocyclic and heterocyclic rings which may be saturated or unsaturated, and which may be mono-, bi- or tri-cyclic, and includes furyl, isoxazolyl, oxazolyl, pyridyl, pyrimidyl, pyrrolyl, thiazolyl, thienyl, triazolyl, morpholinyl, piperazinyl, piperidyl, tetrahydropyranyl, phenyl, cyclohexyl, cyclopentyl, cyclohexanyl, naphthyl, quinolinyl, indolyl, norbornyl, azabicyclooctyl, adamantyl 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.
The term "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 of the invention conform to formula I:
Figure imgf000010_0001
1 0 'X wherein R , R , R , A, D, B, m, n, x, and y are defined as described above.
In one embodiment, variable m is 0 and variable n is 2. In further embodiments A is selected from the group consisting of CH2 and O.
In other embodiments, D is -(CR5R6)Z-. In still other embodiments, z is preferably 1. In other embodiments, each of R5 and R6 is H.
In still other embodiments,
Figure imgf000010_0002
epresents a piperidine ring.
Further embodiments provide for R3 being a 5- to 7-membered ring that is optionally substituted by 1-3 R1 groups. The ring may contain one or more heteroatoms independently selected from the group consisting of N, O and S. Li certain embodiments, R is phenyl that is optionally substituted by 1-3 R1.
Additional embodiments provide for compounds of formula I wherein m is 0, n is 2, and A is CH2 or O. In these embodiments, R1 is selected from the group consisting of H, F, Cl, Br, I, nitro, Ci-6-alkyl, C1-6-alkylhalo, C1-6-alkylhalo, OQ-6-alkylhalo, aryl, C1-6-alkylenearyl, and OCi-6-alkylenearyl, while R is selected from H and Ci-6-alkyl.
When compounds of the present invention contain one or more chiral centers, those compounds 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.
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. By the same token, the present invention encompasses tautomers of the compounds of formula I.
In addition, certain compounds of the present invention may exist in solvated forms, such as hydrated, as well as in unsolvated forms. Thus, the present invention encompasses all such solvated forms of the compounds of formula I.
Also within the scope of the invention are 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 possible as well 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, in particular an acid addition salt such as a hydrochloride, hydrobromide, phosphate, acetate, fumarate, maleate, tartrate, citrate, methanesulphonate orjy-toluenesulphonate.
Specific examples of the present invention include the following compounds, their pharmaceutically acceptable salts, hydrates, solvates, optical isomers, and combinations thereof:
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Preparation of Compounds
Compounds of the present invention can be prepared by various synthetic processes. 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.
Some starting materials for preparing compounds of the present invention are available from commercial chemical sources. Other starting compounds, as described below, are readily prepared from available precursors using straightforward transformations that are well known in the art.
Within these general guidelines, the compounds of formula I generally can be prepared according to the process illustrated in Scheme 1. Variables in Scheme 1 are as defined for formula I hereinabove unless otherwise specified.
Scheme 1
Figure imgf000027_0001
LG in Scheme 1 above represents a leaving group that is capable of being displaced by precursor (ii). Suitable leaving groups are well known in the art and thus include but are not limited to chloride, bromide, and sulphate esters such as mesylate and tosylate. Precursor (i) can be prepared by a number of processes as described in more detail below. An exemplary process may be selected and/or adapted according to the particular structural features of a given precursor (i). Scheme 2 illustrates one exemplary process for making precursor (i). Thus, 6-Fluoro-2-methyl-l,2,3,4-tetrahydroquinoline was formylated using the mixed formic/acetic anhydride. This amide was then regioselectively nitrated with nitronium tetrafluoroborate. The formyl group was removed under basic hydrolytic conditions, and the nitro group was reduced to the aniline using hydrogen gas and palladium on carbon. The benzimidazole ring system was finally formed using chloroacetic acid or an equivalent in the presence of a mineral acid catalyst.
Scheme 2
Figure imgf000027_0002
Figure imgf000027_0003
Scheme 3 illustrates another method for synthesizing precursor (i). 4-fluoroaniline was N- protected with a Boc-protecting group. This group was then used to direct ortho-lithiation, and subsequent trapping with 3-chloro-l-iodopropane provided 6-fluoro-3,4-dihydro-2H- quinoline-1-carboxylic acid tert-butyl ester. The Boc group was replaced with a formyl group, and the synthesis proceeded in an analogous fashion to Scheme 2.
Scheme 3
Figure imgf000028_0001
Figure imgf000028_0002
Figure imgf000028_0003
Scheme 4 illustrates another method for synthesizing precursor (i). Amino-3-nitrophenol was reacted with 1,2-dibromoethane under basic conditions to produce 5-nitro-3,4-dihydro-2H- 1,4-benzoxazine. The synthesis then proceeded as outlined in Scheme 2.
Scheme 4
Figure imgf000028_0004
Scheme 5 illustrates another method for synthesizing precursor (i). 8-Nitroquinolone was reduced using hydrogen gas and platinum oxide catalyst. The resultant product was cyclized to the benzimidazole using chloroacetic acid or an equivalent in the presence of a mineral acid catalyst.
Scheme 5
Figure imgf000029_0001
Scheme 6 illustrates another method for synthesizing precursor (i). This scheme is analogous to Scheme 2.
Scheme 6
Figure imgf000029_0002
Precursor (ii) in Scheme 1 can be obtained from commercial sources or otherwise synthesized by using well known synthetic methodologies. In general, precursor (ii) can be prepared, if necessary, by the route depicted in Scheme 7 below. Boc-protected 4-piperidone was transformed to the vinyl triflate, which could in turn be converted into the cyclic boronate ester using standard conditions. This intermediate underwent Suzuki reaction conditions with various aryl halides in the presence of a palladium catalyst, yielding the final compounds after deprotection.
Scheme 7
Figure imgf000030_0001
Pharmaceutical Composition
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 association 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. Medical Use
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, more particularly as positive allosteric modulators, of the mGluR2 receptor, and are useful in therapy, in particular for the treatment of neurological and psychiatric disorders associated with glutamate dysfunction in an animal.
More specifically, the neurological and psychiatric disorders 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 Chorea, 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, a compound 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, intrathoracically, intravenously, epidurally, intrathecally, intracerebroventricularly and by injection into the joints. In preferred 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 standardization 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.
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. The 1H and 13C NMR spectra were recorded either on Bruker 300, Bruker DPX400 or
Varian +400 spectrometers operating at 300, 400 and 400 MHz for ^H 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 IcV 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 ran 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. 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 ah, 1992, Neuron, 8:169; Miller et ah, 1995, J. Neuroscience, 15:6103; Balazs, et ah, 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+]; 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 Gs 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 370C overnight. Cells were loaded with 6μM fluo-3 acetoxymethylester (Molecular Probes, Eugene Oregon) for 60 min. at room temperature. All assays were performed in a buffer containing 126mM NaCl, 5mM KCl, ImM MgCl2, ImM CaCl2, 2OmM Hepes, 0.06μM DCG-IV (a Group II mGluR selective agonist), supplemented with 1.0mg/ml D-glucose and 1.0mg/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 (O.OlμM to 30μM 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 (0.2μM) 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 niGluR2 receptor activation. The allosteric activator activity of compounds at the human mGluR2 receptor was 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 [ 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 μg protein) were incubated with test compound (3nM to 300μM) for 15 min. at room temperature prior to the addition of 1 μM glutamate, and incubated for 30 min at 3O0C in 500 μ\ assay buffer (20 mM HEPES, 10OmM NaCl, 1OmM MgCl2), containing 30μM GDP and O.lnM [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 (1OmM 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.
The following abbreviations are used in the examples:
BOC tert-butoxycarbonyl
BSA Bovine Serum Albumin
CCD Charge Coupled Device
CRC Concentration Response Curve
DBU l,8-diazabicyclo[5.4.0]undec-7-ene
DCM dichloromethane
DHPG 3,5-dihydroxyphenylglycine;
DIBAL diisobutylaluminum hydride
DMF N,N-dimethylformamide
DMSO dimethyl sulfoxide
EDTA Ethylene Diamine Tetraacetic Acid
Et3N triethylamine
EtOAc Ethyl acetate
FLIPR Fluorometric Imaging Plate reader
GC/MS gas chromatograph coupled mass spectroscopy
GHEK Human Embryonic Kidney expressing Glutamate Transporter
HEPES 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid (buffer)
IP3 inositol triphosphate
MCPBA 3-chloroperbenzoic acid
MeOH methanol
NMP N-Methylpyrrolidinone
NMR nuclear magnetic resonance
PCC pyridinium chlorochromate ppm parts per million
RT room temperature
SPE solid phase extraction
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin layer chromatography Generally, the compounds of the present invention were active in the assays described herein at concentrations (or with ECs0 values) of less than 10 μM. Preferred compounds of the invention have EC50 values of less than 1 μM; more preferred compounds of less than about 100 nJVL For example, the compounds of Examples 26.55, 26.56, 26.65, 26.69, and 28.1 have EC50 values of 0.37, 1.58, 0.08, 0.23, and 1.11 μM, respectively.
Preparation of Intermediate Compounds
Preparation of Precursor (i)
Example 1.1: Tert-butyl (4-fluorophenyl) carbamate
Figure imgf000038_0001
To a solution of (4-fluorophenyl)-amine (5g, 45mmol) in THF (200ml), di-tert-butyl dicarbonate (10.8g, 50 mmol) was added. The resulting mixture was fluxed for 3 h.. After removal of solvents, the residue was dissolved in EtOAc and washed with 10% citric acid, water, brine, and dried over anhydrous sodium sulphate, filtered and concentrated to get a brown solid. This brown solid was washed with hexanes to provide a white solid (8.5g, 89%). 1H NMR (300 MHz, CDCl3): δ 7.28-7.40 (m, 2H), 6.97-7.02 (m, 2H), 6.50 (s, IH), 1.53 (s, 9H).
In a similar fashion, the following compounds were made:
Example 2.1: Tert-butyl-6-fluoro-3,4-dihydroquinoline-l(2H)-carboxylate
Figure imgf000038_0002
A 1.6M solution of tert-butyllithium in pentane (37 ml, 59.17 mmol) was added in a dropwise fashion to a solution of tert-butyl (4-fluorophenyl) carbamate (5 g, 23.67mmol) in anhydrous THF (200ml, Argon atmosphere) at -78°C. After 15 min at -78°C, the reaction was allowed to warm to -20°C where it was maintained for 3 h.. The resulting solution of ortho lithiated compound was then quenched at -20°C with the solution of l-chloro-3iodopropane (2.8ml, 26.03 mmol) in anhydrous THF (20ml), stirred a further 20 min, and then refluxed overnight. The reaction mixture was then quenched with water and the product was extracted with DCM. The organic layer was washed with brine and dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 5% to 10% ethyl acetate/hexanes) provided a yellow oil (2.85g, 48%). 1H NMR (300 MHz, CDCl3): δ 7.55-7.59 (m, IH), 6.69-6.78(m, 2H), 3.62-3.66 (m, 2H), 2.68 (t, 2H), 1.83- 1.87 (m, 2H), 1.48 (s, 9H).
In a similar fashion, the following compound was made:
Figure imgf000039_0002
Example 3.1 : 6-fluoro-2-methyl-3,4-dihydroquinoline-l(2H)-carbaldehyde
Figure imgf000039_0001
The mixture of formic acid (28ml, 726.3 mmol) and acetic anhydride (23 ml, 242 mmol) was added to 6-fluoro-2-methyl-l,2,3,4-tetrahydroquinoline (4 g, 24.2 mmol) dropwise. The resulting mixture was stirred at 60 °C for 1 h.. After removal of solvents, the residue was basified with 3N aqueous sodium hydroxide solution. The product with extracted with dichloromethane and the organic layer was washed with brine and dried over anhydrous Na2- SO4. The solvent was removed under reduced pressure to afford a red oil (4.8 g, yield 103%). 1H NMR (300 MHz, CDCl3): δ 8.53 (s, IH), 6.99-7.04 (m, IH), 6.82-6.87 (m, 2H), 4.73 (t, IH), 2.50-2.70 (m, 2H), 2.02-2.10 (m, IH), 1.58-1.64 (m, IH), 1.12 (d, 3H).
In a similar fashion, the following compound was made:
Figure imgf000040_0003
Example 4.1: 6-fluoro-3,4-dihydroquinoline-l(2H)-carbaldehyde
Figure imgf000040_0001
Tert-butyl-6-fluoro-3,4-dihydroquinoline-l(2H)-carboxylate (2.8 g, 11.16 mmol) was dissolved in the mixture of TFA and DCM (10ml, v/v=l :1) and stirred overnight. After removal of solvents, the mixture of formic acid (10.5 ml, 278.9 mmol) and acetic anhydride (8.4 ml, 89.3 mmol) was added to the residue dropwise. The resulting mixture was stirred at 60 °C for 1 h.. After removal of solvents, the residue was basified with 3N aqueous sodium hydroxide solution. The product with extracted with dichloromethane and the organic layer was washed with brine, dried over anhydrous Na2SO4. The solvent was removed under reduced pressure to afford a red oil (1.4 g, yield 70%). 1H NMR (300 MHz, CDCl3): δ 8.61(s, IH), 6.99-7.04 (m, IH), 6.72-6.84 (m, 2H), 3.68-3.72 (m, 2H), 2.71 (t, 2H), 1.81-1.89 (m, 2H).
In a similar fashion, the following compounds were made:
Figure imgf000040_0004
Example 5.1 : 6-fluoro-2-methyl-8-nitro-3,4-dihydroquinoline-l(2H)-carbaldehyde
Figure imgf000040_0002
To a suspension of nitronium tetrafluoroborate (8.2g, 58.7 mmol) in dichloromethane (50 ml), the solution of 6-fluoro-2-methyl-3,4-dihydroquinoline-l(2H)-carbaldehyde(4.8g, 24.84 mmol) in dichloromethane (20ml) was added dropwise at 0 0C. The reaction was stirred at 0 °C for 2 h., then poured into ice-water (40 ml), the product was extracted with dichloromethane. The combined organic layers were washed with water, brine, and dried over anhydrous Na2SO4. The solvent was removed in vacuo to afford a yellow solid (5.8g, 97%), confirmed by GC/MS.
In a similar fashion, the following compounds were made:
Figure imgf000041_0002
Example 6.1: 6-fluoro-2-methyl-8-nitro-l52,3,4-tetrahydroquinoline
Figure imgf000041_0001
A suspension of 6-fluoro-2-methyl-8-nitro-3,4-dihydroquinoline-l(2H)-carbaldehyde (5.8g, 24.3mmol) in the mixture of ethanol and 10% NaOH aqueous solution (100ml, v/v=l:l) was refluxed over night. After cooled to room temperature, the reaction mixture was diluted with water (200ml), the product was extracted with dichloromethane. The organic layer was washed with brine, dried over anhydrous Na2SO4. The solvent was removed in vacuo to afford a red solid (5.Og, 98%), confirmed by GC/MS.
In a similar fashion, the following compounds were made:
Figure imgf000041_0003
Figure imgf000042_0004
Preparation of Precursors
Figure imgf000042_0001
Example 7.1: 4-Chloro-3,5-dinitro-benzoic acid methyl ester
Figure imgf000042_0002
To a solution of 4-chloro-3,5-dinitro-benzoic acid (2g, 7.8mmol) in methanol (10 mL) was added concentrated H2SO4 (1 mL) dropwise. The reaction mixture was refluxed for 5 h.. The reaction mixture was then cooled to room temperature and kept in O0C bath for 30 min. Product was as obtained off-white solid after filtration (2.1Og, quantitative yield). 1H NMR (300 MHz, CDCl3): δ 8.62 (s, 2H), 4.05 (s, 3H).
Example 8.1: 3,3-Dimethyl-5-nitro-3,4-dihydro-2H-benzo[l,4]oxazine-7-carboxylic acid methyl ester
Figure imgf000042_0003
To a solution of 2-amino-2-methyl-propan-l-ol (1.44g, 16.16mmol) in methanol (15 mL) was added 4-Chloro-3,5-dinitro-benzoic acid methyl ester (2.1g, 8.08mmol). The resulting reaction mixture was refluxed for 45 min. After the reaction mixture was cooled to room temperature, sodium methoxide (1.22g, 22.58mmol) was added slowly, and then the reaction mixture was refluxed for 45 min. The reaction mixture was cooled to room temperature and ice-water was added. The precipitate obtained by filtration was purified on silica gel eluting with 10-20% ethyl acetate in hexane to give the product as yellow solid (900mg, 42%). 1H NMR (300 MHz, CDCl3): δ 8.54 (d, IH), 8.12 (br, IH), 7.64 (d, IH), 3.91 (s, 5H), 1.4 (s, 6H).
Preparation of Precursors
Figure imgf000043_0001
Example 9.1: 2-Amino-5-chloro-3-nitro-phenol
Figure imgf000043_0002
The solution of 2-amino-3-nitro-phenol (3g, 19.46mmol) and N-chlorosuccinimide (3.12g, 23.35mmol) in acetonitrile (100 mL) was refluxed for 3 h.. The reaction mixture was concentrated and the residue was dissolved in ethyl acetate. The mixture was washed with water and brine. The organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo to give product as red solid (3.7g, quantitative yield). 1H NMR (300 MHz, CDCl3): δ 7.55 (d, IH), 6.83 (d, IH).
Example 10.1: 7-Chloro-5-nitro-4H-benzo[l,4]oxazin-3-one
Figure imgf000043_0003
2-Amino-5-chloro-3-nitro-phenol (3.7g, 19.46mmol) was dissolved in acetonitrile (100 mL). Bromo-acetyl chloride (3.37g, 21.40mmol) was added followed by potassium carbonate (6.72g, 48.65mmol). The reaction mixture was refluxed overnight. After removal of the solvent, the residue was partitioned between ethyl acetate and water. The aqueous phase was extracted with ethyl acetate (2x). The combined organic phases were washed with brine, dried over anhydrous sodium sulfate and concentrated in vacuo. The crude residue was purified on silica gel eluting with 20-50% ethyl acetate in hexane to give product as brown solid (2.4g, 54%). 1H NMR (300 MHz, CDCl3): δ 7.94 (d, IH), 7.31 (d, IH), 4.74 (s, 2H).
Example 11.1: 5-Amino-7-chloro-4H-benzo[l,4]oxazin-3-one
Figure imgf000044_0001
Lithium aluminum hydride (1.07g, 22.4mmol) was added to the suspension of 7-chloro-5- nitro-4H-benzo[l,4]oxazin-3-one (1.2g, 5.3mmol) in THF (30 mL). The reaction mixture was stirred at room temperature for overnight. The reaction mixture was then quenched with water, the aqueous phase was extracted with ethyl acetate; combined organic phases were washed with water and brine, dried over anhydrous sodium sulfate and concentrated in vacuo. The crude residue was purified on silica gel eluting with 60% ethyl acetate in hexane and 2% methanol in ethyl acetate to give the product as red oil (460mg, 47%). 1H NMR (300 MHz, CDCl3): δ 6.39 (d, IH), 6.33 (d, IH), 4.17 (m, 2H), 3.53 (br, 2H), 3.41 (m, 2H), 3 (br, IH).
Example 12.1: 6-fluoro-2-methyl-l,2,3,4-tetrahydroquinoline-8-amine
Figure imgf000044_0002
To a solution of 6-fluoro-2-methyl-8-nitro- 1,2,3 ,4-tetrahydroquinoline (4.5g, 21.43mmol) in ethanol (100ml), 10% Pd/carbon (600mg) was added, followed by charging a hydrogen-filled balloon on the top of flask. The reaction was stirred at room temperature for 36 h.. The reaction mixture was filtered through diatomaceous earth and filtrate was condensed to get a colorless oil (3.7g, 96%), confirmed by GC/MS.
In a similar fashion, the following compounds were made:
Figure imgf000044_0003
Figure imgf000045_0004
Example 13.1 : l,2,3,4-tetrahydroquinoline-8-amine
Figure imgf000045_0001
A suspension of 8-nitroquinoline (500 mg, 2.87 mmol) and PtO2 (16 mg, 0.072 mmol) in glacial acetic acid (5 ml) was stirred with H2-balloon on the top for three days. After removal of acetic acid, the residue was partitioned between dichloromethane and saturated sodium bicarbonate solution. The aqueous layer was back-extracted with DCM, and combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, elution with 10% ethyl acetate/ DCM) provided a red oil (120 mg, 24%), confirmed by GC/MS.
In a similar fashion, the following compounds were made:
Figure imgf000045_0003
Example 14.1: 2-(chloromethyl)-8-fluoro-4-methyl-5,6-dihydro-4H-imidazo[4,5,l- ijjquinoline
Figure imgf000045_0002
To a suspension of 6-fluoro-2-methyl-l,2,3,4-tetrahydroquinoline-8-amine (3.7g, 20.6mmol) in 2-chloro-l,l,l-trimethoxyethane (25ml), cone. HCl (3ml) was added. The resulting solution was stirred over night. The reaction mixture was diluted with dichloromethane and basified with saturated sodium bicarbonate solution. The aqueous layer was back-extracted with dichloromethane, and the combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 60% ethyl acetate/hexanes to 5% 2M NH3 in methanol/ (60% ethyl acetate/hexanes)) provided a yellow solid (2.8g, 56%). H NMR (300 MHz, CDCl3): δ 7.17(dxd, IH), 6.78(d, IH), 4.73-4.78 (m, 3H), 2.81-2.99 (m, 2H), 2.07-2.13 (m, 2H), 1.42 (d, 3H).
In a similar fashion, the following compounds were made:
Figure imgf000046_0001
Figure imgf000047_0003
Preparation of Precursor (ii)
Figure imgf000047_0001
Example 15.1 : Tert-butyl-4-{[(trifluoromethyl)sulfonyl]oxy}-3,6-dihydropyridine-l(2H)- carboxylate
Figure imgf000047_0002
To a solution of N5 N-diisopropyl amine (4.2 ml, 30 mmol) in anhydrous THF (130 ml), the solution of n-BuLi in pentane (15 ml, 30mmol) was added in a dropwise fashion at 00C. 15 min later, the solution of tert-butyl-4-oxopiperidine-l-carboxylate (4.98 g, 25 mmol) in anhydrous (60 ml) was added to the reaction at -780C in a dropwise fashion. 30 min later, a solution of 2,2,2-trifluoro-N-phenyl-N-[(trifluoromethylsulfonyl]ethanesulfonamide (9.8 g, 27.5 mmol) was added to the reaction mixture. After 1 h., the reaction was allowed to warm up to room temperature and stirred 3 h.. The reaction mixture was quenched with saturated sodium bicarbonate solution (100 ml), and the product was extracted with EtOAc. The organic layer was washed with water, a brine solution, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 5% to 10% ethyl acetate/hexanes) provided off-white solid (5.88 g, 75%). 1H NMR (300 MHz, CDCL3): 5.70 (br, IH), 4.00 (br, 2H), 3.58 (t, 2H), 2.39 (br, 2H), 1.42 (s, 9H).
Example 16.1: Tert-butyl-4-[ (l,l,2,2,-tetramethyl)-boronate ester]-3,6-dihydropyridine- 1 (2H)-carboxylate
Figure imgf000048_0001
To a solution of tert-butyl-4-{[(trifluoromethyl)sulfonyl]oxy}-3,6-dihydropyridine-l(2H)- carboxylate (5.88 g, 18.65 mmol) in dioxane (60 ml), bis(pinacolate) diboron (5.16 g, 20.51 mmol), [l,r-bis(diphenylphosphino)-ferrocene] dichloropalladiurn (910 mg, 1.12 mmol) and sodium acetate (4.6 g, 55.95 mmol) were added. The resulting mixture was stirred at 80 0C overnight. After removal of solvent, the residue was partitioned between EtOAc and water. The organic layer was washed with water, a brine solution, dried over anhydrous Na2- SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 10% to 20% ethyl acetate/hexanes) provided off-white solid (1.75 g, 35%). 1H NMR (300 MHz, CDCL3): 6.40 (br, IH), 3.89 (br, 2H), 3.77 (t, 2H), 2.17 (br, 2H), 1.40 (s, 9H), 1.20 (s, 12H).
Example 17.1: Tert-Butyl 4-(4-fluorophenyl)-3,6-dihydropyridine-l(2H)-carboxylate
Figure imgf000049_0001
The solution of l-fluoro-4-iodobenzene (463.2 mg, 0.0.74 mmol) in DMF (15 ml) was degassed, and back-filled with Argon. Tert-butyl-4-[(l,l,2,2,-tetramethyl)-boronate ester]- 3,6-dihydropyridine-l(2H)-carboxylate (250 mg, 0.81 mmol), [l,l'-bis(diphenylphosphino)- ferrocene] dichloropalladium (60 mg, 0.074 mmol) and potassium carbonate (305 mg, 2.2 mmol)) were added to the solution. The resulting mixture was stirred over night at 110 0C, then was poured into water and extracted three times with ethyl acetate. The combined organic layers were washed with a brine solution, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 10% to 20% ethyl acetate/hexanes) provided a dark-green oil (153 mg, 75%). 1H NMR (300 MHz, CDCL3): 7.32-7.35 (m, 2H), 6.99-7.05 (m, 2H), 5.90 (br, IH), 4.07 (br, 2H), 3.64 (t, 2H), 2.50 (br, 2H), 1.51 (s, 9H).
hi a similar fashion, the following compounds were made:
Figure imgf000049_0002
Figure imgf000050_0002
Example 18.1: 4-(2,5-difluorophenyl)-piperidine
Figure imgf000050_0001
To a solution of tert-Butyl 4-(2,5.di-fluorophenyl)-3,6-dihydropyridine-l(2H)-carboxylate (53 mg, 0.179 mmol), palladium oxide (20 mg) was added, followed by charging a hydrogen- filled balloon on the top of flask. The reaction was stirred at room temperature overnight. The reaction mixture was filtered through diatomaceous earth and filtrate was condensed to get a colorless oil, which was further stirred in TFA/DCM (2 ml, v/v=l:l) overnight. Removal of solvents provided a yellow gum (40 mg, 81%), confirmed with GC/MS.
In a similar fashion, the following compounds were made:
Figure imgf000051_0003
Figure imgf000051_0002
Example 19.1: Tert-butyl-4-allylpiperidine-l-carboxylate
Figure imgf000051_0001
To a solution of tert-butyl-4-(2-oxoethyl)piperidine-l-carboxylate (2.13 g, 9.37 mmol) in acetonitrile (30 ml), 1, 8-diazabicyclo[5,4,0]undec-7-ene (2.85 g, 18.74 mmol) and methyltriphenyl phosphonium bromide (6.69 g, 18.74 mmol) were added. The reaction was refluxed overnight and diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 10% to 20% ethyl acetate/hexanes) provided a yellow oil (1.91 g, 91%). 1H NMR (300 MHz, CDCL3): 5.62-5.80 (m, IH), 4.94-4.99 (m, IH), 4.93 (s, IH), 4.07 (br, 2H), 2.62 (br, 2H), 1.96 (t, 2H), 1.59-1.63 (m, 2H), 1.41 (s, 9H), 1.08-1.12 (m, IH), 0.96-1.06 (m, 2H).
In a similar fashion, the following compounds were made:
Figure imgf000052_0002
Example 20.1 : Tert-butyl-4- [3 -(4-fluorophenyl)propyl]piperidine-l -carboxylate
Figure imgf000052_0001
Tert-butyl-4-allylpiperidine-l -carboxylate (500 mg, 2.22 mmol) was degassed and back-filled with Argon. 9-BBN (0.5 M in THF, 4.44 ml, 2.66 mmol) was added through a syringe. The mixture was stirred at 600C for 1 h.. After cooled to room temperature, it was added to the mixture of l-bromo-4-fluorobenzene (466 mg, 2.66 mmol), potassium carbonate (460 mg, 3.33 mmol), and [l,r-bis(diphenylphosphino)-ferrocene] dichloropalladium (54 mg, 0.067 mmol) in DMF (10 ml) and water (0.1 ml). The resulting mixture was stirred at 850C for 40 h.. After cooled down to room temperature, it was diluted with water and the product was extracted with EtOAc three times. The combined organic layers were washed with water, brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 10% to 20% ethyl acetate/hexanes) provided a yellow oil (520 mg, 73%). 1H NMR (300 MHz, CDCL3): 7.07-7.12 (m, 2H), 6.90-6.96 (m, 2H), 4.07 (br, 2H)5 2.52-2.70 (m, 4H), 156-1.64 (m, 4H), 1.44 (s, 9H), 1.19- 1.39 (m, 3H), 086-1.06 (m, 2H).
hi a similar fashion, the following compounds were made:
Figure imgf000053_0003
Figure imgf000053_0001
Example 21.1: Tert-butyl-4-(2-bromoethyl)piperidine-l -carboxylate
Figure imgf000053_0002
To a solution of tert-butyl-4-(2-hydroxyethyl)piperidine-l -carboxylate (1.5 g, 6.54 mmol) and tetrabromide carbon (3.25 g, 9.81 mmol) in DCM (20 ml), the solution of triphenylpliosphine (1.72 g, 6.54 mmol) was added slowly. The reaction was stirred overnight and diluted with hexanes (50 ml), washed with water, brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 10 to 30% ether/hexanes) provided a yellow oil (1.67 g, 88%). 1H NMR (300 MHz, CDCL3): 4.07 (br, 2H), 3.35 (br, 2H), 2.60 (br, 2H), 1.69-1.72 (m, 2H), 1.56-1.60(m, 3H), 1.36 (s, 9H), 0.86-1.06 (m, 2H).
Example 22.1: Tert-butyl-4-[2-(4-fluorophenoxy)ethyl]piperidine-l-carboxylate
Figure imgf000054_0001
To a solution of tert-butyl-4-(2-bromoethyl)piperidine-l-carboxylate (600 mg, 2.05 mmol) in acetone (30 ml), 4-fluorophenol (230 mg, 2.05 mmol), potassium carbonate (1.12 g, 8.2 mmol) and tetrabutylammonium iodide (45 mg, 0.123 mmol) was added. The resulting mixture was refluxed overnight. After removal of acetone, the residue was partitioned between water and water. The organic layer was washed with IN NaOH three times, water, brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo to provide a yellow oil (700 mg, 98%). 1H NMR (300 MHz, CDCL3): 6.89-6.94 (m, 2H), 6.75-6.80 (m, 2H), 4.07 (br, 2H), 3.91 (t, 2H), 2.61 (br, 2H), 165-1.68 (m, 5H), 1.43 (s, 9H), 1.02-1.18 (m, 2H).
In a similar fashion, the following compounds were made:
Figure imgf000054_0002
Example 23.1: 6-Fluoro-8-nitro-2,3-dihydro-lH-quinolin-4-one
Figure imgf000055_0001
(i) 3-[(4-fluoro-2-nitrophenyl)amino]propanenitrile
To a stirring solution of (4-fluoro-2-nitrophenyl)amine (1Og, 64.1mmol) in 1,4-dioxane was added acrylonitrile (6.23mL, 96.1mmol) and 40% Triton B in water (0.5mL). The mixture stirred at room temperature overnight and was concentrated. The resulting black solid was dried under vacuum for 3 h., suspended in ether, and stirred vigorously for another 4 h.. The suspension was filtered, washed with ether and dried under vacuum to afford 8.47g (63%) of the title compound as a brown solid. 1H NMR (300 MHz, CDCl3): δ 8.08 (br s, IH), 7.99- 7.95 (dd, IH), 7.37-7.31 (m, IH), 6.89-6.84 (dd, IH), 3.77-3.71 (q, 2H), 2.79-2.75 (t, 2H).
(ii) 3-[(4-fluoro-2-nitrophenyl)amino]propanoic acid
3-[(4-fluoro-2-nitrophenyl)amino]propanenitrile (5.72g, 27.3mmol) was suspended in methanol (50 mL) and 10% sodium hydroxide (50 mL) was added. The mixture refluxed for 2 h., was cooled to room temperature and was concentrated. The resulting slurry was diluted with water (100 mL) and was acidified with 10% hydrochloric acid 100 mL) to pH ~1. The resulting suspension was filtered and washed with water. The resulting aqueous washes were combined, re-acidified, filtered and washed with water. The precipitate was combined and dried under vacuum providing the title compound as an orange solid (4.9Og, 79%). 1H NMR (300 MHz, CDCl3): δ 8.10 (br s, IH), 7.79-7.91 (m, IH), 7.34-7.30 (m, IH), 6.91-6.87 (dd, IH), 3.69-3.67 (m, 2H), 2.82-2.78 (t, 2H).
(iii) 6-fluoro-8-nitro-2,3-dihydroquinolin-4(lH)-one To a flask stirring with Eatons Reagent (11 mL) was added 3-[(4-fluoro-2- mtrophenyl)amino]propanoic acid (0.65g, 2.85mmol). The mixture stirred at 6O0C for 3.5 h. and was then cooled to room temperature. Ice chips were added and the mixture was poured into water. The suspension was filtered and the precipitate was washed with water and dried under vacuum. Purification by column chromatography (silica gel, gradient elution with 5% to 25% ethyl acetate/hexanes) provided the title compound as an orange solid (0.342g, 57%). 1H NMR (300 MHz, CDCl3): δ: 8.19 (br s, IH), 8.17-8.13 (dd, IH), 8.00-8.97 (dd, IH), 3.84- 3.78 (m, 2H), 3.87-3.82 (m, 2H). Example 24.1 : 8-Amino-6-fluoro-2,3-dihydro-lH-quinolin-4-one
Figure imgf000056_0001
A solution of 6-Fluoro-8-nitro-2,3-dihydro-lH-quinolin-4-one in ethyl acetate (25mL) and acetic acid (1OmL) was purged with argon and 10% palladium on carbon was added (200mg). This mixture was stirred for 18 h. at room temperature under hydrogen and was filtered through diatomaceous earth concentrated in vacuo. The crude product was used directly in the next step. 1H NMR (300 MHz, CDCL3): 7.13 (dd, IH), 6.65 (dd, IH), 3.59 (dd, 2H), 2.72 (dd, 2H).
Example 25.1 : 2-Chloromethyl-8-fluoro-6,6-dimemoxy-5,6-dihydro-4H-imidazo[4,5,l- ij]quinoline
Figure imgf000056_0002
To 8-Amino-6-fluoro-2,3-dihydro-lH-quinolin-4-one was added 2-chloro-l,l,l-triethoxy ethane (1OmL) and concentrated hydrochloric acid (0.5mL). The reaction was stirred for 0.5 h. and diluted with dichloromethane (25 ml), and quenched with saturated sodium bicarbonate. The organics were washed with water and brine, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 30 to 60% ethyl acetate/hexanes) and trituration with hexanes afforded a yellow solid (0.722 g, 53% over 2 steps). 1H NMR (300 MHz, CDCL3): 7.38 (dd, IH), 7.22 (dd, IH), 4.84 (s, 2H), 4.39 (dd, 2H), 3.33 (s, 6H), 2.43 (dd, 2H).
Preparation of Final Compounds
Figure imgf000056_0003
Example 26.1 : 8-fluoro-4-methyl-2-[(4-phenylpiperidin-l-yl)methyl]-5,6-dihydro-4H- imidazo[4,5, 1 -ijjquinoline
Figure imgf000057_0001
To a solution of 2-(chloromethyl)-8-fluoro-4-methyl-5,6-dihydro-4H-imidazo[4,5,l- ij]quinoline (23.8 mg, 0.114 mmol ) in acetonitrile (5 ml), 4-phenylpiperidine (27.6 mg, 0.17 mmol) and potassium carbonate (79 mg, 0.57 mmol) were added. The resulting mixture was stirred overnight. Then, the resulting reaction mixture was diluted with water and the product was extracted with EtOAc. The aqueous layer was back-extracted with EtOAc, and the combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. Purification by flash chromatography (silica gel, gradient elution with 80% ethyl acetate/hexanes to 5% 2M NH3 in methanol/ (80% ethyl acetate/hexanes)) provided a brown foam (34.7 mg, 84%). 1H NMR (300 MHz, CDCl3): δ 7.21-7.35 (m, 6H), 6.81-6.84 (m, IH)5 4.97-4.99 (m, IH), 3.85 (s, 2H) 2.94-3.09 (m, 4H), 2.29-2.41 (m, IH), 2.16-2.32 (m, 4H), 1.65-1.86 (m, 3H), 1.50 (d, 3H), 1.15-1.32 (m, IH).
In a similar fashion, the following compounds were made:
Figure imgf000057_0002
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0002
Example 27.1 : 8-Fluoro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-6,6-dimethoxy-5,6- dihydro-4H-imidazo[4,5 , 1 -ij ] quinoline
Figure imgf000084_0001
A solution of 2-Chloromethyl-8-fluoro-6,6-dimethoxy-5,6-dihydro-4H-imidazo[4,5, 1 - ij]quinoline (0.10Og, 0.351mmol), 4-(4-Fluoro-phenyl)-piperidine (0.091g, 0.421 mmol), and potassium carbonate (0.145g, 1.05mmol) in acetonitrile was stirred overnight at room temperature. The mixture was diluted with ethyl acetate, washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. Purification by flash column chromatography (silica gel, gradient elution with 90% ethyl acetate/hexanes to 3% 2M ammonia/methanol in dichloromethane) afforded a light yellow solid (0.124g, 83%). 1H NMR (300 MHz, CDCL3): 7.35 (dd, IH), 7.17 (m, 3H), 6.98 (m, 2H), 4.44 (dd, 2H), 3.85 (s, 2H)5 3.33 (s, 6H), 3.00 (m, 2H), 2.56 (m, IH), 2.40 (m, 2H), 2.26 (m, 2H), 1.73 (m, 4H).
Example 28.1 : 8-Fluoro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-4,5,9a,9b- tetrahydroimidazo[455,l-ij]quinolin-6-one
Figure imgf000085_0001
8-Fluoro-2-[4-(4-fluoro-phenyl)-piρeridin-l-ylmethyl]-6,6-dimethoxy-5,6-dihydro- 4Himidazo[4,55l-ij]quinolme was dissolved in 4mL of TFA (20% in dichloromethane) and stirred for 1 h. at room temperature. The solvent was removed in vacuo and the resulting residue was dissolved in ethyl acetate. The organics were washed with saturated sodium bicarbonate, water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. Purification by flash column chromatography (silica gel, gradient elution with 50% ethyl acetate/hexanes to 2% 2M ammonia/methanol in dichloromethane) afforded a white solid (0.078g, 88%). %). 1HNMR (300 MHz, CDCL3): 7.64 (dd, IH), 7.47 (dd, IH), 7.18 (m, 2H), 7.00 (m, 2H), 4.73 (t, 2H), 3.91 (s, 2H), 3.14 (t, 2H), 3.00 (m, 2H), 2.56 (m, IH), 2.32 (m, 2H), 1.87 (m, 2H), 1.75 (m, 2H).
Example 29.1: 4-Fluoro-l-[4-(4-fluoro-phenyl)-piperidm-l-ylmethyl]-8,9-dihydro-7H-2,7,9a- triaza-benzo[cd]azulen-6-one
Figure imgf000085_0002
To a flask stirring with methane sulfonic acid (2mL) in a cold water bath was slowly added 8-Fluoro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-4,5,9a,9b-tetrahydroimidazo[4,5,l- ij]quinolin-6-one (0.050g, 0.131mmol) and sodium azide (0.01 Ig, 0.170mmol). After stirring at room temperature for 3 h., the mixture was cooled by adding ice chips and neutralized with saturated sodium bicarbonate. The aqueous mixture was then extracted with ethyl acetate and the organics were washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. Purification by flash column chromatography (silica gel, elution with 2% 2M ammonia/methanol in dichloromethane) and trituration with dichloromethane afforded a light yellow solid (0.02Og, 38%). 1H NMR (300 MHz, CDCL3): 7.87 (dd, IH), 7.62 (dd, IH), 7.32 (m, IH), 7.17 (m, 2H), 6.99 (m, 2H), 5.31 (s, 2H), 4.62 (br, 2H), 3.86 (s, 2H), 3.82 (m, 2H), 2.95 (m, 2H), 2.54 (m, IH), 2.29 (m, 2H), 1.86 (m, 2H) 1.70 (m, 2H). Example 30.1 : (i) [4R]- and (U) [4S]-8-Fluoro-4-methyl-2-{[4(4-trifluoromethyl- phenyl)piperizin-l-yl]niethyl}-5,6-dihydro-4H-imidazo[4,5,l-ij]quinoline
Figure imgf000086_0001
Racemic 8-fluoro-4-niethyl-2-{[4(4-trifluoromethyl-phenyl)piperizin-l-yl]metliyl}-5,6- dihydro-4H-imidazo[4,5,l-ij]quinoline was separated into its constituent enantiomers using HPLC on a 21.4 mm x 250 mm Chiralcel OJ column. Elution was effected with 25% EtOH in petroleum ether at 15 mL/min.

Claims

WE CLAIM:
1. A compound of formula (I) :
Figure imgf000087_0001
wherein
A is selected from the group consisting of CR R , NR5, O, S5 SO and SO2;
B is selected from the group consisting of CH and N;
D is selected from the group consisting of NH, N-C1-6-alkyl, and -(CR 5T R) 6N V, wherein one of the -CR5R6- groups maybe replaced by -C(O)-, NH, or NC1-6-alkyl;
L is selected from the group consisting of a direct bond and -(CR5R6)w-5 wherein when L is -(CR5R6V: (i) B-L may be unsaturated, or two adjacent carbon atoms may form part of a cyclopropyl ring; or (ii) one or two CR5R6 groups may be replaced with O, S, or NR5;
Figure imgf000087_0002
represents a ring selected from the group consisting of azetidine and a 5- to 7- membered ring, which may be unsaturated, wherein the ring may be substituted by one or more R4;
R1, in each instance, is selected from the group consisting of H, F, Cl, Br, I, OH, CN, nitro, C1-6-alkyl, OC1-6-alkyl, C1-6-alkylhalo, OC1-6-alkylhalo, C2-6-alkenyl, OC2-6- alkenyl, C2-6-alkynyl, OC2-6-alkynyl, C3-8-cycloalkyl, C1-6-alkylene-C3-8-cycloalkyl, OC0-6-alkylene-C3-8-cycloalkyl, aryl, heteroaryl, Ci-6-alkylenearyl, C1-6- alkyleneheteroaryl, OC1-6-alkylenearyl, OCi-6-alkyleneheteroaryl, C1-6- alkyleneheterocycloalkyl, (CO)R5, (CO)OR5, Ci-6-alkylene0R5, OC2-6-alkyleneOR5, C1-6-alkylene(CO)R5, OC1-6-alkylene(CO)R5, C1-6-alkylenecyano, OC2-6- alkylenecyano, C0-6-alkyleneNR6R7, OC2-6-alkyleneNRcR7, C,-6-alkylene(CO)NR6R7, OCi-6-alkylene(CO)NR6R7, C0-6-alkyleneNR6(CO)R7, OC2-6-alkyleneNR6(CO)R7, C0- 6-alkyleneNR6(CO)NR6R7, C0-6-alkyleneSO2R5, OC2-6-alkyleneSO2R5, C0-6- alkylene(SO2)NR6R7, OC2-6-alkylene(SO2)NR6R7, C0-6-alkyleneNR5(SO2)R7, OC2-6- alkyleneNR6(SO2)R7, C0-6-alkyleneNR6(SO2)NR6R7, OC2.6-alkyleneNR6(SO2)NR6R7, (CO)NR6R7 and SO3R5, wherein any cyclic group may be further substituted with one or more R2 groups;
R2 and R4, in each instance, are independently selected from the group consisting of H, F, Cl, Br, I, CN, nitro, hydroxy, oxo, C1-6-alkyl, OC1-6-alkyl, C1-6-alkylhalo, OC1-6- alkylhalo, and C0-6-alkyleneNR5R6;
R3 is a 5- to 12-membered ring system that is optionally substituted by up to three R groups, wherein the ring system may contain one or more heteroatoms independently selected from the group consisting of N, O and S;
R5 is selected from the group consisting of H, C1-6-alkyl, aryl, C3-s-cycloalkyl, C1-6- alkylenearyl and Q-e-alkylene-Q-g-cycloalkyl, wherein any cyclic group may be further substituted with one or more independently-selected R groups;
R6 and R7 are independently selected from the group consisting of H and C1-6-alkyl;
R8 and R9 are independently selected from the group consisting of H, -O-(CH2)2-O- and - O-(CH2)3-O-;
m and n are integers independently selected from the group consisting of O, I5 2, 3 and 4, with the proviso that m and n cannot simultaneously be O;
x andy are integers independently selected from the group consisting of 1, 2, and 3; and w and z are integers independently selected from the group consisting of I5 2, 3, 4, 5, and 6; or a pharmaceutically-acceptable salt, hydrate, solvate, isoform, tautomer, optical isomer, or combination thereof.
2. The compound according to claim 1, wherein m is 0 and n is 2.
3. The compound according to claim 1, wherein A is selected from the group consisting OfCH2 and O.
4. The compound according to claim 1, wherein D is -(CR5R6)Z-.
5. The compound according to claim 4, wherein z is 1.
6. The compound according to claim 4, wherein each of R5 and R6 is H.
7. The compound according to claim 1, wherein
Figure imgf000089_0001
represents piperidine.
8. The compound according to claim 1, wherein R3 is a 5- to 7-membered ring that is optionally substituted by 1-3 R1 groups, wherein the ring may contain one or more heteroatoms independently selected from the group consisting of N, O and S.
9. The compound according to claim 8, wherein R3 is phenyl that is optionally substituted by 1-3 R1 groups.
10. The compound according to claim 1, wherein m is 0 and n is 2;
A is CH2 or O;
R1 is selected from the group consisting of H, F, Cl, Br, I, nitro, C1-6-alkyl, C1-6- alkylhalo, C1-6-alkylhalo, OC1-6-alkylhalo, aryl, Ci-6-alkylenearyl, and OC1-6- alkylenearyl; and R2 is selected from H and C1-6-alkyl.
11. A compound selected from the group consisting of :
8-fluoro-4-methyl-2-[(4-phenylpiperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ijjquinoline; 8-fluoro-4-methyl-2-{4-[3-(4-fluorophenyl) propyl]piperidin-l-ylmethyl}-5,6-diliydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2- {4-[2-(4-fluorophenoxy) ethyl]piperidin-l-ylmethyl} -5,6-dihydro-4H- irnidazo [4, 5 , 1 -ij ] quinoline;
2-[(4-phenylpiperidin-l-yl)methyl]-4,5-dihydroimidazo[l,5,4-de][l,4]benzoxazine; 2- {4-[3-(4-fluorophenyl) propyl] piperidin-l-ylmethyl}-4,5-dihydroimidazo[l, 5,4- de] [ 1 ,4]benzoxazine;
2-{4-[2-(4-fluorophenoxy) ethyl]piperidin-l-ylmethyl}-4,5-dihydroimidazo[l,5,4- de] [ 1 ,4]benzoxazine;
2-{4-[3-(3-fluoro-5-(trifluoromethyl)-phenyl) propyl] piperidin-l-ylmethyl}-4,5- dihydroimidazo [ 1 , 5 ,4-de] [ 1 ,4]benzoxazine;
2- [(4-phenylpiperidin- 1 -yl)methyl] -5 ,6-dihydro-4H-imidazo [4, 5 , 1 -ij] quinoline; 2-{4-[3-(4-fluorophenyl) propyl] piperidin-l-ylmethyl}-5,6-dihydro-4H-imidazo[4,5,l- ij]quinoline;
2-{4-[2-(4-fluorophenoxy) ethyl]piperidin-l-ylmethyl}-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-methoxy-2-[(4-phenylpiperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l-ij]quinolme; 8-methyl-2- [(4-phenylpiperidin- 1 -yl)methyl] -5 ,6-dihydro-4H-imidazo [4,5 , 1 -ij ] quinoline; 8-methyl-2-{4-[3-(4-fluorophenyl) propyl] piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-methyl-2-{4-[2-(4-fluorophenoxy) ethyl]piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-methyl-2- {4-[3-(3-fluoro-5(trifluoromethyl)-phenyl) propyl] piperidin-l-ylmethyl}-5,6- dihydro-4H-imidazo[4,5, 1 -ij] quinoline;
8-methyl-2-[(4-benzylpiperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l-ij]quinoline; 8-methyl-2-{[4-(4-bromophenyl)piperidin-l-yl]methyl}-5,6-dihydro-4H-imidazo[4,5,l- ij]quinoline;
8-methyl-2-{[4-(4-cyanophenyl)piperidin-l-yl]methyl}-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-methyl-2- { [4-(4-chlorophenyl)piperidin-l -yl]methyl} -5 ,6-dihydro-4H-imidazo [4,5, 1 - ij]quinoline; 8-methyl-2-[(4-phenoxypiperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l-ij]quinoline; 8-methyl-2- { [4-(3 -hydroxyphenyl)piperidin- 1 -yljmethyl} -5 ,6-dihydro-4H-imidazo[4,5, 1 - ij]quinoline;
8-fluoro-4-methyl-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-4-methyl-5,6-dihydro-4H- imidazo[4,5, 1 -ij]quinoline;
8-fluoro-4-methyl-2-[4-(4-trifluoromethylphenyl)-piperidin-l-ylraethyl]-5,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2-[4-(4-bromo-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2-[4-(4-cyano-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5 , 1 -ij] quinoline;
8-fluoro-4-methyl-2-[4-(4-methylphenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4, 5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2-[4-(4-methoxyphenyl)-piperidm-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(2-methoxyphenyl)-piperidin- 1 -ylmethyl] -5 ,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2-[4-(2-methylphenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-2- [(4-phenylpiperidin- 1 -yl)methyl] -5 ,6-dihydro-4H-imidazo [4, 5 , 1 -ij ] quinoline; 8-fluoro-2- [(4-benzylpiperidin- 1 -yl)methyl] -5 ,6-dihydro-4H-imidazo [4, 5 , 1 -ij ] quinoline; 8-fluoro-2-[4-(3-phenyl propyl)piperidin-l-ylmethyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-fluoro-2-{4-[3-(4-fluorophenyl) propyl]piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-2-{4-[2-(4-fluorophenoxy) ethyl]piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-2-{[4-(4-fluorophenyl)piperidm-l-yl]methyl}-5,6-dihydro-4H-iniidazo[4,5,l- ij] quinoline;
8-fluoro-2- { [4-(4-trifluoromethyl-phenyl)piperidin- 1 -yl]methyl} -5 ,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-chloro-2- { [4-(4-fluorophenyl)piperidin- 1 -yl]methyl} -5 ,6-dihydro-4H-imidazo[4,5 , 1 - ij] quinoline; S-chloro-2- { [4-(4-trifluoromethyl-phenyl)piperidin- 1 -yl]raethyl} -5 ,6-dihydro-4H- itnidazo[4,5,l-ij]quinoline;
S-chloro-2-{4-[3-(4-fluorophenyl) propyl]piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(2-trifluoromethylphenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(3-fluoro-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(3-trifluoromethylphenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2- { [4-(4-fluorophenyl)-3,6-dihydropyridin- 1 (2H)-yl]methyl} -5 ,6-dihydro- 4H-imidazo[4,5 , 1 -ijjquinoline;
8-fluoro-2-{[4-(4-fluorophenyl)-3,6-dihydropyridin-l(2H)-yl]methyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-chloro-2- {[4-(4-fluorophenyl)-3,6-dihydropyridin-l (2H)-yl]methyl} -5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-{[4-(2,5-difluorophenyl)-3,6-dihydropyridin-l(2H)-yl]methyl}-5,6- dihydro-4H-imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-2-{[4-(2,5-difluorophenyl)-3,6-dihydropyridin-l(2H)-yl]methyl}-5,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-chloro-2- {[4-(2,5-difluorophenyl)-3,6-dihydropyridin-l (2H)-yl]methyl} -5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-fluoro-4-methyl-2-[4-(4-chlorophenyl)-piperidin-l-ylniethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(3-chlorophenyl)-piperidm-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5, 1 -ij ] quinoline;
8-fluoro-4-methyl-2-[4-(3,5-bis(trifluoromethyl)-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro- 4H-imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2-[4-benzylphenyl)-piperidin-l-ylmethyl]-5,6-diliydro-4H-imidazo[455,l- ij] quinoline;
8-fluoro-4-methyl-2-[4-(3-methoxyphenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5, 1 -ij]quinoline; 8-fluoro-4-methyl-2-[4-(3-methylphenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(2-chlorophenoxy)-ρiperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-fluoro-4-methyl-2-{[4(4-fluorophenyl)piperizin-l-yl]methyl}-5,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2-{[4(4-trifluoromethyl-phenyl)piperizin-l-yl]methyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-fluoro-4-methyl-2-{[4(2,4-difluorophenyl)piperizin-l-yl]methyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(2-fluoro-phenyl)-piperidin-l-ylniethyl]-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-fluoro-4-methyl-2-[4-phenoxy-piperidin-l-ylmethyl]-5,6-diliydro-4H-iniidazo[4,5,l- ij] quinoline;
8-fluoro-4-metliyl-2-[3-phenyl-piperazm-l-ylmethyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-fluoro-4-methyl-2-{[4-(2,5-difluorophenyl)piperidin-l-yl]methyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-{[4-(4-bromophenyl)-3,6-dihydropyridin-l(2H)-yl]metliyl}-5,6-dihydro- 4H-imidazo[4,5, 1 -ij] quinoline;
8-fluoro-2-{[4-(4-bromophenyl)-3,6-dihydropyridin-l(2H)-yl]methyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
8-fluoro-4-methyl-2- { [4-(4-trifluoromethoxyphenyl)-3 ,6-dihydropyridin- 1 (2H)-yl]methyl} - 5,6-dihydro-4H-imidazo[4,5, 1 -ij]qumoline;
8-fluoro-2- { [4-(4-trifluoromethoxyphenyl)-3 ,6-dihydropyridin- 1 (2H)-yl]methyl} -5,6- dihydro-4H-imidazo[4,5,l-ij]quinoline;
2-[(4-(4-fluorophenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l-ij]quinoline; 2-[(4-(4-trifluoromethylphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
2-[(4-(4-fluorophenylpiperidm-l-yl)methyl]-4,5-dihydroimidazo[l,5,4-de][l,4]benzoxazine; 2-[(4-(4-trifluoromethylphenylpiperidin-l-yl)methyl]-4,5-dihydroimidazo[l,5,4- de] [ 1 ,4]benzoxazine; [4R]-8-fluoro-4-methyl-2-[(4-phenylpiperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
[4S]-8-fluoro-4-methyl-2-[(4-phenylpiperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-fluoro-4-methyl-2-{[4-(4-trifluoromethoxyphenyl)piperidin-l-yl]methyl}-5,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-2-{[4-(4-trifluoromethoxyphenyl)piperidin-l-yl]methyl}-5,6-dihydro-4H- imidazo[4,5 , 1 -ij]quinoline;
8-chloro-2-[(4-(2-trifluoromethylphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H- imidazo [4,5 , 1 -ij ] quinoline;
8-chloro-2-[(4-(3-trifluoromethylphenyl)piperidin-l-yl)metliyl]-5,6-dihydro-4H- imidazo [4, 5 , 1 -ij ] quinoline;
8-chloro-2- [(4-(4-chlorophenyl)pip eridin- 1 -yl)methyl] -5 ,6-dihydro-4H-imidazo [4,5,1- ij] quinoline;
8-chloro-2-[(4-(3-chlorophenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-chloro-2-[(4-(2-fluorophenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-chloro-2- [(4-(3 -fluorophenyl)piperidin- 1 -yl)methyl] -5 ,6-dihydro-4H-imidazo [4, 5 , 1 - ij] quinoline;
8-chloro-2-[(4-(2-methylphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-chloro-2-[(4-(3-methylphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-chloro-2-[(4-(4-methylplienyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ijjquinoline;
8-chloro-2-[(4-(3-methoxyphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ijjquinoline;
8-chloro-2-[(4-(4-methoxyphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij] quinoline;
8-fluoro-2- {[4-(3-fluorophenyl)piperidin- 1 -yl]methyl} -5,6-dihydro-4H-imidazo[4,5, 1 - ij] quinoline; 8-chloro-2-[(4-(2-methoxyphenyl)piperidin-l-yl)methyl]-5,6-dihydro-4H-imidazo[4,5,l- ij]quinoline;
8-fluoro-4-methyl-2-[4-(2,4-difluoro-phenyl)-piperidin-l-ylmethyl]-5,6-dihydiO-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2-[4-(4-fluoro-3-trifluoromethyl-phenyl)-piperidin-l-ylmethyl]-5,6- dihydro-4H-imidazo[4,5,l-ij]quinoline;
8-fluoro-2-[4-(4-fluoro-3-trifluoromethyl-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H- imidazo [4, 5 , 1 -ij ] quinoline;
8-fluoro-4-methyl-2- {[4-(3-trifluoromethoxyphenyl)piperidin-l -yljmethyl} -5,6-dihydro-4H- imidazo[4,5, 1 -ijjquinoline;
8-fluoro-2- {[4-(3-trifluoromethoxyphenyl) piperidin-l-yl]methyl} -5,6-dihydro-4H- imidazo[4,5, 1 -ij]quinoline;
8-fluoro-4-methyl-2- { [4-(2-trifluoromethoxyphenyl)piperidin- 1 -yljmethyl} -5 ,6-dihydro~4H- imidazo[4,5, 1 -ij] quinoline;
8-fluoro-2-{[4-(2-trifluoromethoxy phenyl) piperidin-l-yl]methyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ijjquinoline;
8-fluoro-4-methyl-2-{4-[2-(3,4-difluorophenoxy) ethyl] piperidm-l-ylmethyl}-5,6-dihydro- 4H-imidazo[4,5, 1 -ijjquinoline;
8-fluoro-2-{4-[2-(3,4-difluorophenoxy) ethyl] piperidm-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ij] quinoline;
2-{4-[2-(3,4-difluorophenoxy) ethyl] piperidin-l-ylmethyl}-4,5-dihydroimidazo[l,5,4- de] [ 1 ,4]benzoxazine;
8-chloro-2-{4-[2-(3,4-difluorophenoxy) ethyl] piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-4-methyl-2- {4-[2-(3,5-difluorophenoxy) ethyl] piperidin-l-ylmethyl}-5,6-dihydro- 4H-imidazo [4,5 , 1 -ij ] quinoline;
8-fluoro-2-{4-[2-(3,5-difluorophenoxy) ethyl] piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ijjquinoline;
2- {4- [2-(3 , 5 -difluorophenoxy) ethyl] piperidin- 1 -ylmethyl} -4,5 -dihydroimidazo [1,5,4- de] [ 1 ,4]benzoxazine;
8-chloro-2-{4-[2-(3,5-difluorophenoxy) ethyl] piperidin- 1 -ylmethyl} -5,6-dihydro-4H- imidazo [4, 5 , 1 -ij ] quinoline; 8-fluoro-2- {4-[2-(4-fluorophenyl) ethyl]piperidin- 1 -ylmethyl } -5 ,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-chloro-2-{4-[2-(4-fluorophenyl) ethyl]piperidin-l-ylmethyl}-5,6-dihydro-4H- imidazo[4,5, 1 -ij]quinoline;
2-{4-[2-(4-fluorophenyl) ethyl] piperidin-l-ylmethyl}-4,5-dihydroimidazo[l,5,4- de] [ 1 ,4]benzoxazine;
8-fluoro-2- {[4-(2,4-difluorophenyl)piperidin- 1 -yljmethyl} -5,6-dihydro-4H-imidazo[4,5, 1 - ijjquinoline;
2-[(4-2,4-difluorophenyl piperidin-l-yl)methyl]-4,5-dihydroimidazo[l,5,4- de] [ 1 ,4]benzoxazine;
7-chloro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H-imidazo[4,5,l- ijjquinoline;
7,8-difluoro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-5,6-dihydro-4H-imidazo[4,5,l- ijjquinoline;
2- [4-(4-fluoro-phenyl)-piperidin- 1 -ylmethyl] -3,3 -dimethyl-3 ,4-dihydro-5 -oxa- 1 ,2a-diaza- acenaphthylene-7-carboxylic acid methyl ester;
2-[4-(3-fluoro-phenyl)-piperidin-l-ylmethyl]-3,3-dimethyl-3,4-dihydro-5-oxa-l,2a-diaza- acenaphthylene-7-carboxylic acid methyl ester;
2-{4-[2-(4-fluoro-phenyl)-ethyl]-piperidin-l-ylmethyl}-3,3-dimethyl-3,4-dihydro-5-oxa-l,2a- diaza-acenaphthylene-7-carboxylic acid methyl ester;
7-chloro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-3,4-dihydro-5-oxa-l,2a-diaza- acenaphthylene;
7-chloro-2-[4-(3-fluoro-phenyl)-piperidin-l-ylmethyl]-3,4-dihydro-5-oxa-l,2a-diaza- acenaphthylene;
7-chloro-2- {4-[2-(4-fluoro-phenyl)-ethyl]-piperidin- 1 -ylmethyl} -3,4-dihydro-5-oxa- 1 ,2a- diaza-acenaphthylene;
7-chloro-2-[4-(2,4-difluoro-phenyl)-piperidin-l-ylmethyl]-3,4-dihydro-5-oxa-l,2a-diaza- acenaphthylene;
7-chloro-2-{4-[3-(4-fluoro-phenyl)-propyl]-piperidin-l-ylmethyl}-3,4-dihydro-5-oxa-l,2a- diaza-acenaphthylene;
7-chloro-2- {4-[2-(4-fluoro-phenoxy)-ethyl]-piperidin- 1 -ylmethyl} -3 ,4-dihydro-5-oxa- 1 ,2a- diaza-acenaphthylene; 8-fluoro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-6,6-dimethoxy-5,6-dihydro-4H- imidazo[4,5,l-ij]quinoline;
8-fluoro-2-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-4;,5,9a,9b-tetrahydroimidazo[4,5Jl- ij]quinolin-6-one;
4-fluoro-l-[4-(4-fluoro-phenyl)-piperidin-l-ylmethyl]-8,9-dihydro-7H-2,7,9a-triaza- benzo[cd] azulen-6-one;
[4R]-8-fluoro-4-methyl-2-{[4(4-trifluoromethyl-phenyl)piperizin-l-yl]methyl}-5,6-dihydro- 4H-imidazo [4,5 , 1 -ij ] quinoline; and
[4S]-8-fluoro-4-methyl-2-{[4(4-trifluoromethyl-phenyl)piperizin-l-yl]methyl}-5,6-dihydro- 4H-imidazo [4,5 , 1 -ij ] quinoline.
12. A pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically acceptable carrier or excipient.
13. 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 claim 1.
14. 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 of formula I in the form of a pharmaceutical composition according to claim 12.
15. The method according to claim 13 or 14, wherein the neurological and psychiatric disorders are selected from the group consisting of 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, AlDS-induced dementia, Alzheimer's disease, Huntington's Chorea, 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, migraine, urinary incontinence, substance tolerance, substance withdrawal, psychosis, schizophrenia, anxiety, mood disorders, circadian rhythm disorders, trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain, tardive dyskinesia, sleep disorders, attention deficit/hyp eractivity disorder, and conduct disorder.
16. The pharmaceutical composition according to claim 12, for use in the prevention and/or treatment of mGluR2 receptor-mediated disorders.
17. A compound according to claim 1 for use in therapy.
18. The compound according to claim 17, for use in the prevention and/or treatment of mGluR2 receptor-mediated disorders.
19. The use of a compound according to claim 1 in the manufacture of a medicament for the use in the prevention and/or treatment of mGluR2 receptor-mediated disorders.
20. A process for the preparation of a compound of formula I according to claim 1 , said process comprising the step of reacting a precursor compound according to formula (i):
Figure imgf000098_0001
with a precursor compound according to formula (ii):
Figure imgf000098_0003
to give the compound according to formula I:
Figure imgf000098_0002
wherein R1, R2, , RR33,, AA,, DD,, LL,, mm,, nn,, xx,, aanndd yy aarree aass ddeeffiinneecd in claim 1, and wherein LG is a leaving group.
PCT/US2006/028165 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators WO2007018998A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EP06787957A EP1912989A2 (en) 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators
US11/996,727 US20080318999A1 (en) 2005-08-05 2006-07-21 Tricyclic Benzimidazoles and Their Use as Metabotropic Glutamate Receptor Modulators
CA002616020A CA2616020A1 (en) 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators
BRPI0614168A BRPI0614168A2 (en) 2005-08-05 2006-07-21 COMPOUND OR A PHARMACEUTICALLY ACCEPTABLE SALT, HYDRATE, SOLVATE, ISOFORM, TAUTOMER, OPTICAL ISOMER OR A COMBINATION THEREOF, PHARMACEUTICAL COMPOSITION, METHOD FOR THE TREATMENT OR PREVENTION OF NEUROLOGICAL AND PSCHIATRIC DISORDERS, USE OF A COMPOUND, AND, PROCESS FOR THE PREPARATION OF A COMPOUND
AU2006279034A AU2006279034A1 (en) 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators
JP2008524995A JP2009503069A (en) 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as modulators of metabolic glutamate receptors
MX2008001152A MX2008001152A (en) 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators.
IL188809A IL188809A0 (en) 2005-08-05 2008-01-16 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators
NO20080475A NO20080475L (en) 2005-08-05 2008-01-25 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US70547105P 2005-08-05 2005-08-05
US60/705,471 2005-08-05

Publications (2)

Publication Number Publication Date
WO2007018998A2 true WO2007018998A2 (en) 2007-02-15
WO2007018998A3 WO2007018998A3 (en) 2007-05-03

Family

ID=37607094

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/028165 WO2007018998A2 (en) 2005-08-05 2006-07-21 Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators

Country Status (17)

Country Link
US (2) US20080318999A1 (en)
EP (1) EP1912989A2 (en)
JP (1) JP2009503069A (en)
KR (1) KR20080035576A (en)
CN (1) CN101268077A (en)
AR (1) AR055100A1 (en)
AU (1) AU2006279034A1 (en)
BR (1) BRPI0614168A2 (en)
CA (1) CA2616020A1 (en)
EC (1) ECSP088128A (en)
IL (1) IL188809A0 (en)
MX (1) MX2008001152A (en)
NO (1) NO20080475L (en)
RU (1) RU2008101923A (en)
TW (1) TW200745112A (en)
UY (1) UY29710A1 (en)
WO (1) WO2007018998A2 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
EP2291078A1 (en) * 2008-05-15 2011-03-09 Merck Sharp & Dohme Corp. Oxazolobenzimidazole derivatives
WO2014072435A1 (en) * 2012-11-08 2014-05-15 Selvita Sa Substituted tricyclic benzimidazoles as kinase inhibitors
US9227985B2 (en) 2013-03-15 2016-01-05 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9290514B2 (en) 2013-07-08 2016-03-22 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
WO2016044130A1 (en) * 2014-09-15 2016-03-24 Incyte Corporation Tricyclic heterocycles for use as bet protein inhibitors
US9309246B2 (en) 2013-12-19 2016-04-12 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9315501B2 (en) 2013-11-26 2016-04-19 Incyte Corporation Bicyclic heterocycles as BET protein inhibitors
US9399640B2 (en) 2013-11-26 2016-07-26 Incyte Corporation Substituted pyrrolo[2,3-c]pyridines and pyrazolo[3,4-c]pyridines as BET protein inhibitors
US9540368B2 (en) 2014-04-23 2017-01-10 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US9975906B2 (en) 2014-05-16 2018-05-22 Shionogi & Co., Ltd. Tricyclic heterocycle derivatives having HIV replication inhibitory effect
US10189832B2 (en) 2016-06-20 2019-01-29 Incyte Corporation Crystalline solid forms of a BET inhibitor
US10329305B2 (en) 2015-10-29 2019-06-25 Incyte Corporation Amorphous solid form of a BET protein inhibitor
US10494380B2 (en) 2015-05-29 2019-12-03 Shionogi & Co., Ltd. Nitrogen-containing tricyclic derivatives having HIV replication inhibitory activity
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE529246C2 (en) * 2005-10-13 2007-06-12 Neurosearch Sweden Ab New disubstituted phenyl-piperidines as modulators of dopamine neurotransmission
EP1963268B1 (en) * 2005-12-07 2010-12-01 NSAB, Filial af NeuroSearch Sweden AB, Sverige Disubstituted phenylpiperidines as modulators of cortical catecholaminergic neurotransmission
TWI417095B (en) * 2006-03-15 2013-12-01 Janssen Pharmaceuticals Inc 1,4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of mglur2-receptors
TW200845978A (en) * 2007-03-07 2008-12-01 Janssen Pharmaceutica Nv 3-cyano-4-(4-tetrahydropyran-phenyl)-pyridin-2-one derivatives
TW200900065A (en) 2007-03-07 2009-01-01 Janssen Pharmaceutica Nv 3-cyano-4-(4-pyridinyloxy-phenyl)-pyridin-2-one derivatives
UA100126C2 (en) 2007-09-14 2012-11-26 Аддекс Фарма С.А. 1',3'-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2h, 1'h-[1, 4'] bipyridinyl-2'-ones
JP5433579B2 (en) 2007-09-14 2014-03-05 ジャンセン ファーマシューティカルズ, インコーポレイテッド. 1,3-disubstituted-4-phenyl-1H-pyridin-2-one
PT2200985E (en) 2007-09-14 2011-07-21 Ortho Mcneil Janssen Pharm 1,3-disubstituted 4-(aryl-x-phenyl)-1h-pyridin-2-ones
CN101861316B (en) * 2007-11-14 2013-08-21 奥梅-杨森制药有限公司 Imidazo[1,2-a]pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
DE102008022221A1 (en) * 2008-05-06 2009-11-12 Universität des Saarlandes Inhibitors of human aldosterone synthase CYP11B2
CA2723729A1 (en) * 2008-05-15 2009-11-19 Merck Sharp & Dohme Corp. Oxazolobenzimidazole derivatives
BRPI0913188A2 (en) * 2008-05-29 2016-01-12 Albany Molecular Res Inc 5-ht3 receptor modulators, manufacturing methods, and use thereof
US20110171134A1 (en) * 2008-06-25 2011-07-14 Iskandar Bermans J (6s)-5-methyltetrahydrofolic acid for therapy of tissue injury
AU2009289784B2 (en) 2008-09-02 2012-03-22 Addex Pharma S.A. 3-azabicyclo[3.1.0]hexyl derivatives as modulators of metabotropic glutamate receptors
WO2010036544A1 (en) * 2008-09-26 2010-04-01 Merck Sharp & Dohme Corp. Oxazolobenzimidazole derivatives
RU2517181C2 (en) 2008-10-16 2014-05-27 Орто-Макнейл-Янссен Фармасьютикалз, Инк. Indole and benzomorpholine derivatives as modulator of metabotropic glutamate receptors
BRPI0921333A2 (en) 2008-11-28 2015-12-29 Addex Pharmaceuticals Sa indole and benzoxazine derivatives as metabotropic glutamate receptor modulators
MY153913A (en) 2009-05-12 2015-04-15 Janssen Pharmaceuticals Inc 7-aryl-1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mglur2 receptors
BRPI1010831A2 (en) 2009-05-12 2016-04-05 Addex Pharmaceuticals Sa 1,2,4-triazolo [4,3-a] pyridine derivatives and their as positive allosteric modulators of mglur2 receptors
KR101753826B1 (en) 2009-05-12 2017-07-04 얀센 파마슈티칼즈, 인코포레이티드 1,2,4-triazolo [4,3-a] pyridine derivatives and their use for the treatment or prevention of neurological and psychiatric disorders
US8541404B2 (en) * 2009-11-09 2013-09-24 Elexopharm Gmbh Inhibitors of the human aldosterone synthase CYP11B2
EP2649069B1 (en) 2010-11-08 2015-08-26 Janssen Pharmaceuticals, Inc. 1,2,4-TRIAZOLO[4,3-a]PYRIDINE DERIVATIVES AND THEIR USE AS POSITIVE ALLOSTERIC MODULATORS OF MGLUR2 RECEPTORS
AU2011328203B2 (en) 2010-11-08 2015-03-19 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
ES2536433T3 (en) 2010-11-08 2015-05-25 Janssen Pharmaceuticals, Inc. 1,2,4-Triazolo [4,3-a] pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
WO2012112791A1 (en) 2011-02-16 2012-08-23 Paloma Pharmaceuticals, Inc. Radiation countermeasure agents
AU2012278976B2 (en) 2011-07-06 2017-05-11 Gilead Sciences, Inc. Compounds for the treatment of HIV
CN102964292A (en) * 2012-11-26 2013-03-13 盛世泰科生物医药技术(苏州)有限公司 Synthesis method of 4-(2-(trifluoromethoxy) phenyl) piperidine
JO3368B1 (en) 2013-06-04 2019-03-13 Janssen Pharmaceutica Nv 6,7-DIHYDROPYRAZOLO[1,5-a]PYRAZIN-4(5H)-ONE COMPOUNDS AND THEIR USE AS NEGATIVE ALLOSTERIC MODULATORS OF MGLUR2 RECEPTORS
JO3367B1 (en) 2013-09-06 2019-03-13 Janssen Pharmaceutica Nv 1,2,4-TRIAZOLO[4,3-a]PYRIDINE COMPOUNDS AND THEIR USE AS POSITIVE ALLOSTERIC MODULATORS OF MGLUR2 RECEPTORS
KR101672096B1 (en) * 2013-09-30 2016-11-02 주식회사 엘지화학 Heterocyclic compound and organic light emitting device comprising the same
HUE045610T2 (en) 2014-01-21 2020-01-28 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
MX2016009471A (en) 2014-01-21 2016-10-13 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use.
US10202353B2 (en) 2014-02-28 2019-02-12 Gilead Sciences, Inc. Therapeutic compounds
WO2015154047A1 (en) * 2014-04-03 2015-10-08 Restorgenex Corporation Novel methods
EP3347361B1 (en) 2015-09-08 2020-08-05 H. Hoffnabb-La Roche Ag Tricyclic pi3k inhibitor compounds and methods of use
WO2017049321A1 (en) 2015-09-17 2017-03-23 Miller Marvin J Benzyl amine-containing heterocyclic compounds and compositions useful against mycobacterial infection
UY37367A (en) 2016-08-19 2018-03-23 Gilead Sciences Inc NEW COMPOUNDS FOR USE IN THE TREATMENT OF A VIRAL INFECTION AND COMPOSITIONS OF THE SAME
EP3752495B1 (en) 2018-02-15 2023-07-19 Gilead Sciences, Inc. Pyridine derivatives and their use for treating hiv infection
AR114631A1 (en) 2018-02-16 2020-09-30 Gilead Sciences Inc METHODS AND INTERMEDIATES FOR PREPARING PYRIDINE COMPOUNDS
KR20210025016A (en) 2018-06-28 2021-03-08 지앙수 헨그루이 메디슨 컴퍼니 리미티드 Conjugated tricyclic heterocycle compound and its therapeutic use
KR20210033492A (en) 2018-07-16 2021-03-26 길리애드 사이언시즈, 인코포레이티드 Capsid inhibitors for the treatment of HIV
EP3873896A4 (en) * 2018-12-14 2022-08-31 Jiangsu Hengrui Medicine Co., Ltd. Tricyclic compounds as sting agonists, and preparation methods and medicinal uses thereof
WO2020151682A1 (en) * 2019-01-23 2020-07-30 成都先导药物开发股份有限公司 Macrocyclic immunomodulator
US20220144826A1 (en) * 2019-03-11 2022-05-12 Collaborative Medicinal Development, Llc Heteroaromatic and Heterobicyclic Aromatic Derivatives for the Treatment of Ferroptosis-Related Disorders
CN116283758B (en) * 2023-03-30 2024-05-24 安徽工业大学 Method for synthesizing N-formyl tetrahydroquinoline from quinoline and product

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002012239A1 (en) * 2000-08-08 2002-02-14 Sanofi-Synthelabo Benzimidazole derivatives, preparation and therapeutic use thereof
WO2002044183A2 (en) * 2000-12-01 2002-06-06 Guilford Pharmaceuticals Inc. Benzoazepine and benzodiazepine derivatives and their use as parp inhibitors
US6548494B1 (en) * 1999-08-31 2003-04-15 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(ADP-ribose) polymerases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6548494B1 (en) * 1999-08-31 2003-04-15 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(ADP-ribose) polymerases
WO2002012239A1 (en) * 2000-08-08 2002-02-14 Sanofi-Synthelabo Benzimidazole derivatives, preparation and therapeutic use thereof
WO2002044183A2 (en) * 2000-12-01 2002-06-06 Guilford Pharmaceuticals Inc. Benzoazepine and benzodiazepine derivatives and their use as parp inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
FERRARIS DANA ET AL: "Design and synthesis of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. Part 4: biological evaluation of imidazobenzodiazepines as potent PARP-1 inhibitors for treatment of ischemic injuries." BIOORGANIC & MEDICINAL CHEMISTRY 15 AUG 2003, vol. 11, no. 17, 15 August 2003 (2003-08-15), pages 3695-3707, XP002415270 ISSN: 0968-0896 *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
EP2291078A1 (en) * 2008-05-15 2011-03-09 Merck Sharp & Dohme Corp. Oxazolobenzimidazole derivatives
EP2291078A4 (en) * 2008-05-15 2011-08-24 Merck Sharp & Dohme Oxazolobenzimidazole derivatives
WO2014072435A1 (en) * 2012-11-08 2014-05-15 Selvita Sa Substituted tricyclic benzimidazoles as kinase inhibitors
US9745299B2 (en) 2012-11-08 2017-08-29 Selvita Sa Substituted tricyclic benzimidazoles as kinase inhibitors
KR101812390B1 (en) 2012-11-08 2017-12-26 셀비타 에스에이 Substituted tricyclic benzimidazoles as kinase inhibitors
AU2013343550B2 (en) * 2012-11-08 2016-09-22 Selvita Sa Substituted tricyclic benzimidazoles as kinase inhibitors
US9227985B2 (en) 2013-03-15 2016-01-05 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US10919912B2 (en) 2013-03-15 2021-02-16 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9938294B2 (en) 2013-03-15 2018-04-10 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10464947B2 (en) 2013-03-15 2019-11-05 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US11498926B2 (en) 2013-03-15 2022-11-15 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9624241B2 (en) 2013-03-15 2017-04-18 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
EA038494B1 (en) * 2013-03-15 2021-09-07 Инсайт Холдингс Корпорейшн Tricyclic heterocycles as bet protein inhibitors
US9533997B2 (en) 2013-07-08 2017-01-03 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9290514B2 (en) 2013-07-08 2016-03-22 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9850257B2 (en) 2013-07-08 2017-12-26 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9918990B2 (en) 2013-11-26 2018-03-20 Incyte Corporation Substituted pyrrolo[2,3-c]pyridines and pyrazolo[3,4-c]pyridines as BET protein inhibitors
US9399640B2 (en) 2013-11-26 2016-07-26 Incyte Corporation Substituted pyrrolo[2,3-c]pyridines and pyrazolo[3,4-c]pyridines as BET protein inhibitors
US9737516B2 (en) 2013-11-26 2017-08-22 Incyte Corporation Bicyclic heterocycles as bet protein inhibitors
US9315501B2 (en) 2013-11-26 2016-04-19 Incyte Corporation Bicyclic heterocycles as BET protein inhibitors
US9309246B2 (en) 2013-12-19 2016-04-12 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US11091484B2 (en) 2013-12-19 2021-08-17 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9777003B2 (en) 2013-12-19 2017-10-03 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US10442803B2 (en) 2013-12-19 2019-10-15 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US10781209B2 (en) 2014-04-23 2020-09-22 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US9957268B2 (en) 2014-04-23 2018-05-01 Incyte Corporation 1H-pyrrolo[2,3,c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US9540368B2 (en) 2014-04-23 2017-01-10 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US11059821B2 (en) 2014-04-23 2021-07-13 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US11702416B2 (en) 2014-04-23 2023-07-18 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US10472358B2 (en) 2014-04-23 2019-11-12 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US9975906B2 (en) 2014-05-16 2018-05-22 Shionogi & Co., Ltd. Tricyclic heterocycle derivatives having HIV replication inhibitory effect
US9834565B2 (en) 2014-09-15 2017-12-05 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
WO2016044130A1 (en) * 2014-09-15 2016-03-24 Incyte Corporation Tricyclic heterocycles for use as bet protein inhibitors
US10618910B2 (en) 2014-09-15 2020-04-14 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
TWI712603B (en) * 2014-09-15 2020-12-11 美商英塞特公司 Tricyclic heterocycles as bet protein inhibitors
US9527864B2 (en) 2014-09-15 2016-12-27 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10227359B2 (en) 2014-09-15 2019-03-12 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
CN111138458A (en) * 2015-05-29 2020-05-12 盐野义制药株式会社 Nitrogen-containing tricyclic derivatives having HIV replication inhibiting effect
US10494380B2 (en) 2015-05-29 2019-12-03 Shionogi & Co., Ltd. Nitrogen-containing tricyclic derivatives having HIV replication inhibitory activity
US10870661B2 (en) 2015-05-29 2020-12-22 Shionogi & Co., Ltd. Nitrogen-containing tricyclic derivatives having HIV replication inhibitory activity
US10858372B2 (en) 2015-10-29 2020-12-08 Incyte Corporation Amorphous solid form of a BET protein inhibitor
US10329305B2 (en) 2015-10-29 2019-06-25 Incyte Corporation Amorphous solid form of a BET protein inhibitor
US10189832B2 (en) 2016-06-20 2019-01-29 Incyte Corporation Crystalline solid forms of a BET inhibitor
US11091480B2 (en) 2016-06-20 2021-08-17 Incyte Corporation Crystalline solid forms of a BET inhibitor
US11377446B2 (en) 2016-06-20 2022-07-05 Incyte Corporation Crystalline solid forms of a BET inhibitor
US10626114B2 (en) 2016-06-20 2020-04-21 Incyte Corporation Crystalline solid forms of a BET inhibitor
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Also Published As

Publication number Publication date
BRPI0614168A2 (en) 2017-07-25
NO20080475L (en) 2008-04-15
JP2009503069A (en) 2009-01-29
CA2616020A1 (en) 2007-02-15
EP1912989A2 (en) 2008-04-23
KR20080035576A (en) 2008-04-23
ECSP088128A (en) 2008-02-20
UY29710A1 (en) 2007-02-28
CN101268077A (en) 2008-09-17
WO2007018998A3 (en) 2007-05-03
US20080318999A1 (en) 2008-12-25
MX2008001152A (en) 2008-04-02
AU2006279034A1 (en) 2007-02-15
TW200745112A (en) 2007-12-16
AR055100A1 (en) 2007-08-08
RU2008101923A (en) 2009-09-10
US20070032469A1 (en) 2007-02-08
IL188809A0 (en) 2008-08-07

Similar Documents

Publication Publication Date Title
EP1912989A2 (en) Tricyclic benzimidazoles and their use as metabotropic glutamate receptor modulators
DK2444399T3 (en) Oxadiazole derivatives and their use as enhancers of metabotropic glutamate receptors - 842
US7799792B2 (en) Metabotropic glutamate receptor oxadiazole ligands and their use as potentiators 841
US7485722B2 (en) Spiro-oxazolidinone compounds and their use as metabotropic glutamate receptor potentiators
US20110053953A1 (en) AZA-Isoindolones and Their Use as Metabotropic Glutamate Receptor Potentiators - 613
US20080312271A1 (en) Azabenzimidazolyl compounds
US20120053165A1 (en) Novel Phenyl Imidazoles and Phenyl Triazoles As Gamma-Secretase Modulators
US20110224231A1 (en) Novel Lactams as Beta Secretase Inhibitors
EP1912939A1 (en) Metabotropic glutamate-receptor-potentiating isoindolones
CA2607874A1 (en) Quinoline derivatives as neurokinin receptor antagonists
US7807706B2 (en) Metabotropic glutamate-receptor-potentiating isoindolones
EP2300484B1 (en) Novel class of spiro piperidines for the treatment of neurodegenerative diseases
US20130150376A1 (en) Novel Sultam Compounds

Legal Events

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

Ref document number: 200680034420.5

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: 565007

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 188809

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2616020

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1020087001611

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 12008500181

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2008010127

Country of ref document: EG

WWE Wipo information: entry into national phase

Ref document number: 2008524995

Country of ref document: JP

Ref document number: MX/a/2008/001152

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2006279034

Country of ref document: AU

Ref document number: 08007124

Country of ref document: CO

Ref document number: 2006787957

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 879/DELNP/2008

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006279034

Country of ref document: AU

Date of ref document: 20060721

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2008101923

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 11996727

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0614168

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080125