WO2008130323A1 - Novel 8-piperazine-tetrahydronaphtalene derivatives for the treatment of pain - Google Patents

Novel 8-piperazine-tetrahydronaphtalene derivatives for the treatment of pain Download PDF

Info

Publication number
WO2008130323A1
WO2008130323A1 PCT/SE2008/050462 SE2008050462W WO2008130323A1 WO 2008130323 A1 WO2008130323 A1 WO 2008130323A1 SE 2008050462 W SE2008050462 W SE 2008050462W WO 2008130323 A1 WO2008130323 A1 WO 2008130323A1
Authority
WO
WIPO (PCT)
Prior art keywords
tetrahydronaphthalen
methylpiperazin
pain
phenyl
compounds
Prior art date
Application number
PCT/SE2008/050462
Other languages
French (fr)
Inventor
Yevgeni Besidski
Ylva Gravenfors
Istvan Macsari
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
Publication of WO2008130323A1 publication Critical patent/WO2008130323A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/135Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/38Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals

Definitions

  • the present invention relates to new compounds, to pharmaceutical composition containing said compounds and to the use of said compounds in therapy.
  • the present invention further relates to processes for the preparation of said compounds and to new intermediates useful in the preparation thereof.
  • Voltage-gated sodium channels are critical elements in the control of electrical excitability of various cell types, including muscle and neuronal cells. In muscle and neuronal cells voltage-gated sodium channels are mainly responsible for the rising phase of the action potential. Voltage-gated sodium channels are composed of a single alpha subunit and one or two beta subunits. There are 10 known alpha subunit proteins, of which nine are functional as an ion channel. The different alpha subunit proteins are herein referenced to as Navl.x, with x being an integer between 1 and 9. This labelling is in accordance with the conventions of the International Pharmacological Association (REF).
  • REF International Pharmacological Association
  • Alpha subunits are large proteins of an approximate weight of 260 kDA ( ⁇ 2000 amino acids), and are functional as voltage-gated sodium channels as monomeric structures.
  • Beta subunits are known at present. Beta subunits are smaller proteins of an approximate weight of 33-36 kDa. Beta subunits can modulate functional expression, as well as the characteristics of channel opening and closing (gating) of alpha subunits.
  • voltage-gated sodium channels are important therapeutic targets: a) the biophysical characteristics of voltage-gated sodium channels, b) the tissue expression pattern of voltage-gated sodium channels, c) evidence from preclinical research, d) the association between several congenital diseases and channelopathies of voltage-gated sodium channels, and e) evidence from the usage of pharmacological agents active at voltage-gated sodium channels in the clinic.
  • a main biophysical characteristic of voltage-gated sodium channels is the fast opening and closing (activation and inactivation) of the channel upon an appropriate voltage stimulus.
  • each of these alpha subunits has a characteristic tissue expression pattern. Tissue-specific up- or down-regulation of the expression of several of the voltage-gated sodium channels in human diseases or preclinical disease models in animals strongly supports a central role for specific voltage-gated sodium channels in distinct diseases.
  • Navl.7 is expressed in human neuromas, which are swollen and hypersensitive nerves and nerve endings that are often present in chronic pain states (Acta Neurochirurgica (2002) 144(8) 803-810). Navl.7 is also expressed in dorsal root ganglion neurons and contributes to the small tetrodoxin (TTX) sensitive component seen in these cells. Navl.7 may thus be a potential pain target in addition to its role in neuroendocrine excitability (EMBO Journal (1995) 14(6) 1084-1090).
  • TTX small tetrodoxin
  • the present invention relates to a novel group of compounds that exhibit NaVl.7 inhibiting activity, and are therefore expected to be useful in the prophylaxis and treatment of different acute and chronic pain conditions.
  • WO 97/34883, WO 99/14212, WO 99/05135 and WO 99/14213 describe compounds for use in treatment of pain.
  • the compounds described in these prior art documents bind to serotonine receptors.
  • the compounds of the present invention have little to none activity towards the serotonine receptor.
  • the compounds of the present invention also are contemplated to have an improved pharmacokinetic profile compared to the compounds in the prior art, including a higher oral bioavailability, a decreased clearance and a decreased volume of distribution. Without being bound to any theory, the difference in pharmacokinetic profile is believed to be due to the fact that the right hand side of the molecule is aromatic in the compounds of the present invention while this is not the case for the known compounds.
  • Ri is phenyl or a heteroaromatic ring containing one or two heteroatoms selected from N, O and S and which may be independently mono- or di-substituted with R 2 and/or R 3,
  • R 2 and R 3 are independently selected from Ci-C 6 alkyl, halogen, CF 3 , OH, Ci-C 3 alkoxy, OCF 3 , SO 3 CH 3 , SO 3 CF 3 , phenyl, phenyl-Ci-C 6 alkyl and Ci-C 6 alkyl phenyl; with the proviso that when Ri is phenyl with one substituent R 2 which is Ci-C 6 alkyl, halogen, or CF 3 , then Ri also has a second substituent R 3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a pharmaceutically acceptable salt thereof.
  • R 2 and R 3 are independently selected from Ci-C 6 alkyl, halogen, CF 3 , OH, Ci-C 3 alkoxy, OCF 3 , SO 3 CF 3 and phenyl; with the proviso that when Ri is phenyl with one substituent R 2 which is Ci-C 6 alkyl, halogen, or CF 3 , then Ri also has a second substituent R 3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a pharmaceutically acceptable salt thereof.
  • R 2 and R 3 are independently selected from Ci-C 6 alkyl, halogen, CF 3 , Ci-C 3 alkoxy, OCF 3 , SO 3 CF 3 , or phenyl.
  • R 2 and R 3 are independently selected from CH 3 , C(CH 3 ) 3 , F, Cl, CF 3 , OCH 3 , OCH 2 CH 3 , OCH 2 (CH 3 ) 2 , OCF 3 , SO 3 CF 3 , or phenyl.
  • Ri is phenyl or a heteroaromatic ring containing one or two heteroatoms selected from N, O and S and which may be mono- or di-substituted with R 2 and/or R 3, wherein R 2 is Ci-C 6 alkyl, halogen, CF 3 , OH, Ci-C 3 alkoxy, OCF 3 , SO 3 CH 3 , SO 3 CF 3 , phenyl, phenyl-Ci-C 6 alkyl, Ci-C 6 alkyl phenyl, R 3 is OH, CF 3 , OCF 3 , halogen, Ci-C 6 alkyl or Ci-C 3 alkoxy; with the proviso that when Ri is phenyl with one substituent R 2 which is Ci-C 6 alkyl, halogen, or CF 3 , then Ri also has a second substituent R 3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a
  • Ci-C 6 means a carbon group having 1, 2, 3, 4, 5 or 6 carbon atoms.
  • alkyl includes both straight and branched chain alkyl groups and may be, but are not limited to methyl, ethyl, n-propyl, i- propyl, n-butyl, i-butyl, s-butyl, t-butyl, n-pentyl, i-pentyl, neo-pentyl, n-hexyl or i-hexyl.
  • the term having 1 to 4 carbon atoms and may be but are not limited to methyl, ethyl, n-propyl, i-propyl or t-butyl.
  • alkoxy refers to radicals of the general formula -O-R, wherein R is selected from a hydrocarbon radical.
  • alkoxy may include, but is not limited to methoxy, ethoxy, propoxy, isopropoxy, butoxy, t-butoxy or isobutoxy.
  • amine or “amino” refers to radicals of the general formula -NRR', wherein R and R' are independently selected from hydrogen or a hydrocarbyl radical.
  • aromatic refers to hydrocarbyl radicals having one or more polyunsaturated carbon rings having aromatic character, (e.g., 4n + 2 delocalized electrons) and comprising 6 up to about 14 carbon atoms.
  • aryl used alone or as suffix or prefix, refers to a hydrocarbon radical having one or more polyunsaturated carbon rings having aromatic character, (e.g., 4n + 2 delocalized electrons) and comprising 5 up to about 14 carbon atoms, wherein the radical is located on a carbon of the aromatic ring.
  • alkylphenyl' refers to a phenyl group which is mono-, di-, or tri-substituted with alkyl such as for example methylphenyl in example 4.
  • halo or halogen refers to fluorine, chlorine, bromine and iodine radicals.
  • heterocycle or “heterocyclic” or “heterocyclic ring” refers to ring-containing monovalent and divalent radicals having one or more heteroatoms, independently selected from N, O, P and S, as part of the ring structure and comprising at least 3 and up to about 20 atoms in the rings preferably 5 and 6 membered rings. Heterocyclic ring may be saturated or unsaturated and contain one or more double bonds.
  • heteroaryl refers to heterocyclic monovalent and divalent radicals having aromatic character.
  • heterocyclic monovalent and divalent radicals having aromatic character may be, but is not limited to oxazolyl, isoxazol, furyl, thiophenyl, pyrrolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, indolyl, indazolyl, pyridonyl or benzothienyl.
  • Heteroaryl may also be quinolinyl, quinoxalinyl, naphthyridinyl, isoquinolinyl or thiazolyl.
  • hydrocarbyl refers to any structure comprising only carbon and hydrogen atoms up to 14 carbon atoms.
  • a group R 3 substituted with a group SO 3 CF 3 is an R 3 substituted with trifiuoromenthanesulfonic acid.
  • mammal includes any of various warm-blooded vertebrate animals of the class Mammalia, including but not limited to humans, generally characterized by a covering of hair on the skin.
  • the present invention relates to any one of the specific compounds mentioned above.
  • the present invention relates to the compounds of formula I or Ia as hereinbefore defined as well as to pharmaceutical acceptable salts thereof.
  • Salts for use in pharmaceutical compositions will be pharmaceutically acceptable salts, but other salts may be useful in the production of the compounds of formula I or Ia.
  • a suitable pharmaceutically acceptable salt of the compounds of the invention is, for example, an acid-addition salt, for example a salt with an inorganic or organic acid.
  • a suitable pharmaceutically acceptable salt of the compounds of the invention is an alkali metal salt, an alkaline earth metal salt or a salt with an organic base.
  • the compounds of the present invention may also exists as solvents, solvated hydrates or cocrystals.
  • the compounds of the invention may exhibit tautomerism. All tautomeric forms and mixtures thereof are included within the scope of the invention.
  • the compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism.
  • Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation.
  • the various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques.
  • the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation, or by derivatisation, for example with a homochiral acid followed by separation of the diastereomeric esters by conventional means (e.g. HPLC, chromatography over silica). All stereoisomers are included within the scope of the invention.
  • Another object of the invention relates to processes (a) or (b) for the preparation of compounds of general Formula I and salts thereof.
  • the reaction may be carried out using a suitable acylating reagent such as an acyl chloride, in a suitable solvent such as dichloromethane, chloroform, toluene or acetonitrile at a temperature between -20 0 C and reflux.
  • a suitable acylating reagent such as an acyl chloride
  • a suitable solvent such as dichloromethane, chloroform, toluene or acetonitrile
  • a suitable base may be an organic amine base such as pyridine, 2,6-lutidine, collidine, triethylamine, N,N-diisopropylethylamine, morpholine, 7V-methylmorpholine, diazabicyclo[5.4.0]undec-7-ene or tetramethylguanidine or an alkali metal or an alkaline earth metal carbonate or hydroxide such as sodium carbonate, potassium carbonate, calcium carbonate, sodium hydroxide or potassium hydroxide.
  • the reaction may be aided by the presence of 4-dimethylaminopyridine.
  • the transformation may be performed using a suitable activating reagent such as 2-(1H- benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate or N,N'- carbonyldiimidazole with a suitable base such as triethylamine or N ,N- diisopropylethylamine.
  • a suitable activating reagent such as 2-(1H- benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate or N,N'- carbonyldiimidazole with a suitable base such as triethylamine or N ,N- diisopropylethylamine.
  • the reaction may be performed in a suitable solvent such as dimethylformamide, acetonitrile or dichloromethane at a temperature between -20 0 C and reflux.
  • Protection and deprotection of functional groups may take place before or after any of the reaction steps described hereinbefore.
  • the compounds of the invention exhibit voltage-gated sodium channel inhibiting activity, especially Navl .7 blocking activity, for example as demonstrated in the test described below.
  • the present invention relates to the compounds of formula I or Ia, which inhibit any sodium channel.
  • Modulation of voltage-gated sodium channels by pharmacological or genetical tools points to a central role for distinct voltage-gated sodium channels in several disease models.
  • a mouse line has been generated which through advanced molecular biology technologies eliminates the functional expression of Navl.7 in DRG neurons that express Navl.8 (Proceedings of the National Academy of Sciences USA (2004) 101(34) 12706-12711). This mouse line shows greatly reduced pain responses in several pain behaviour models.
  • Herpes-vector mediated knockdown of Navl.7 in primary afferents of wildtype mice results in a decrease in inflammatory hyperalgesia (Human Gene Therapy (2005) 16(2) 271-277).
  • Antagonists of NaV channels have been shown to be useful for treating a variety of conditions, including acute and chronic nociceptive, visceral, inflammatory, central and peripheral neuropathic pain. More specifically, modulators of NaV activity are currently used or being tested in the clinic as anaesthetics, including local anaesthetics (Pain (2000) 87(1) 7-17), neuropathic pain reliefers (European Journal of Pain (2002) 6(Supplement 1) 61-68), acute pain reliefers (The Cochrane Database of Systematic Reviews (2005) 3), chronic pain reliefers (Pharmacotherapy (2001) 21(9) 1070-1081), inflammatory pain reliefers (Proceedings of the National Academy of Sciences USA (1999) 96(14) 7645- 7649), headache reliefers (Headache (2001) 41(Supplement 1) S25-S32).
  • the compounds of the invention are thus expected to be useful in both the prophylaxis and the treatment of a condition which is effected or facilitated by inhibition of voltage-gated sodium channels, in particular pain, such as acute and chronic pain disorders including but not limited to widespread pain, localized pain, nociceptive pain, inflammatory pain, central pain, central and peripheral neuropathic pain, central and peripheral neurogenic pain, central and peripheral neuralgia, low back pain, postoperative pain, visceral pain, pelvic pain, allodynia, anesthesia dolorosa, causalgia, dysesthesia, fibromyalgia, hyperalgesia, hyperesthesia, hyperpathia, ischemic pain, sciatic pain, pain associated with cystitis, including but not limited to interstitial cystitis, pain associated with multiple sclerosis, pain associated with arthritis, pain associated with osteoarthritis, pain associated with rheumatoid arthritis and pain associated with cancer.
  • pain such as acute and chronic pain disorders including but not limited to widespread pain, localized pain,
  • the compounds of the present invention may be administered alone or in combination with other compounds, especially therapeutically active compounds.
  • the compounds of the present invention may for example be combined with one or more of the following therapeutically active compounds: proton pump inhibitors such as omeprazole, lansoprazole, rabeprazole, tentorpazole, pantoprazole, esomeprazole, revaprazan or sorprazan.
  • proton pump inhibitors such as omeprazole, lansoprazole, rabeprazole, tentorpazole, pantoprazole, esomeprazole, revaprazan or sorprazan.
  • one embodiment of the invention relates to a combination wherein a compound of formula (I) or (Ia) or a pharmaceutically acceptable salt, solvate or in vivo hydrolysable ester, solvates, hydrated solvates, hydrates or co crystals thereof, or a pharmaceutical composition or formulation comprising a compound of formula (I) or (Ia) is administered concurrently, simultaneously, sequentially or separately with another pharmaceutically active compound or compounds selected from the following:
  • antidepressants such as agomelatine, amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, pro trip tyline, ramelteon, reboxetine, robalzotan, sertraline, sibutramine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof, (ii) atypical antipsychotics including for example quetiapine and pharmaceutically active isomer(s) and metabolite(s)
  • antipsychotics including for example amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutylpiperidine, pimozide, prochlorperazine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, ziprasidone
  • anxiolytics including for example alnespirone, azapirones,benzodiazepines, barbiturates such as adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, quazepam, reclazepam, tracazolate, trepipam, temazepam, triazolam, uldazepam, zolazepam and equivalents and pharmaceutically active
  • anticonvulsants including for example carbamazepine, valproate, lamotrogine, gabapentin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • Alzheimer's therapies including for example donepezil, memantine, tacrine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • Parkinson's therapies including for example deprenyl, L-dopa, Requip, Mirapex,
  • MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • migraine therapies including for example almotriptan, amantadine, bromocriptine, butalbital, cabergoline, dichloralphenazone, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pramipexole, rizatriptan, ropinirole, sumatriptan, zolmitriptan, zomitriptan, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • (ix) stroke therapies including for example abciximab, activase, NXY-059, citicoline, crobenetine, desmoteplase,repinotan, traxoprodil and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • urinary incontinence therapies including for example darafenacin, falvoxate, oxybutynin, propiverine, robalzotan, solifenacin, tolterodine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • neuropathic pain therapies including for example gabapentin, lidoderm, pregablin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • nociceptive pain therapies such as celecoxib, etoricoxib, lumiracoxib, rofecoxib, valdecoxib, diclofenac, loxoprofen, naproxen, paracetamol and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • GABAb modulators such as baclofen, and equivalents and pharmaceutically active salts and metabolite(s) thereof.
  • insomnia therapies including for example agomelatine, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, etomidate, glutethimide, halazepam, hydroxyzine, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, ramelteon, roletamide, triclofos,secobarbital, zaleplon, Zolpidem and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • mood stabilizers including for example carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, verapamil, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • Such combination products employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active compound or compounds within therapeutically active dosage ranges and/or the dosage described in the publication reference.
  • One embodiment of the invention relates to compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, for use in therapy.
  • Another embodiment relates to the use of compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, in the manufacture of a medicament for treatment of pain.
  • a further embodiment relates to the use of compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, in the manufacture of a medicament for treatment of acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain.
  • One embodiment relates to a method of treatment of pain, or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain in a patient suffering from, or at risk of, said disease, which comprises administering to the patient a therapeutically effective amount of a compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, or a pharmaceutical composition comprising said compounds.
  • An agent for the treatment of pain, or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain which comprises as active ingredient a compounds of formula I or Ia as defined above , or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, or a pharmaceutical composition comprising said compounds.
  • compositions will normally be administered orally, subcutaneously, intravenously, intraarterially, transdermally, intranasally, by inhalation, or by any other parenteral route, in the form of pharmaceutical preparations comprising the active ingredient either as a free base or a non-toxic organic or inorganic acid addition salt, in a pharmaceutically acceptable dosage form.
  • the compositions may be administered at varying doses.
  • One embodiment relates to a pharmaceutical composition
  • a pharmaceutical composition comprising as active ingredient a therapeutically effective amount of the compound of formula I or Ia as defined above, in association with one or more pharmaceutically acceptable diluents, excipients and/or inert carriers.
  • Another embodiment relates to said pharmaceutical composition according, for use in the treatment of pain or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain.
  • Suitable daily doses of the compounds of the invention in therapeutic treatment of humans are about 0.005 to 25.0 mg/kg body weight at oral administration and about 0.005 to 10.0 mg/kg body weight at parenteral administration.
  • Example of ranges of daily doses of the compounds of the invention in therapeutic treatment of humans are about 0.005 to 10.0 mg/kg body weight at oral administration and about 0.005 to 5.0 mg/kg body weight at parenteral administration.
  • Compounds of the invention may also have the advantage that they may be more efficacious than, be less toxic than, have a broader range of activity than, be more potent than, be longer acting than, produce fewer side effects than, be more easily absorbed than, or that they may have other useful pharmacological properties over, compounds known in the prior art.
  • Microwave irradiation was performed in a Creator TM , Initiator TM or Smith Synthesizer TM Single -mode microwave cavity producing continuous irradiation at 2450 MHz.
  • Mass spectra were recorded on one of the following instruments: a Perkin-Elmer SciX API 150ex spectrometer; a VG Quattro II triple quadrupole; a VG Platform II single quadrupole; or a Micromass Platform LCZ single quadrupole mass spectrometer (the latter three instruments were equipped with a pneumatically assisted electrospray interface (LC- MS)).
  • a Perkin-Elmer SciX API 150ex spectrometer a VG Quattro II triple quadrupole
  • a VG Platform II single quadrupole a Micromass Platform LCZ single quadrupole mass spectrometer
  • Prep-HPLC Preparative chromatography was run on Waters auto purification HPLC with a diode array detector. Column: XTerra MS C8, 19 x 300 mm, 10 ⁇ m. Gradient with acetonitrile/0.1 M ammonium acetate in 5 % acetonitrile in MiIIiQ Water, typically run from 20% to 60% acetonitrile, in 13 min. Flow rate: 20 mL/min. Alternatively, purification was achieved on a semi preparative Shimadzu LC-8A HPLC with a Shimadzu SPD-IOA UV- vis. -detector equipped with a Waters Symmetry ® column (C 18, 5 ⁇ m, 100 mm x 19 mm).
  • NMR spectra were recorded on a Varian Unity+ 400 NMR Spectrometer, operating at 400 MHz for proton and 100 MHz for carbon- 13, and equipped with a 5 mm BBO probe with Z-gradients; or on a Bruker av400 NMR spectrometer operating at 400 MHz for proton and 100 MHz for carbon-13, and equipped with a 3 mm flow injection SEI 1 HZD- 13 C probehead with Z-gradients, using a BEST 215 liquid handler for sample injection; or on a Bruker DPX400 NMR spectrometer, operating at 400 MHz for proton and 100 MHz for carbon-13, and equipped with a 4-nucleus probe with Z-gradients.
  • Rotamers may or may not be denoted in spectra depending upon ease of interpretation of spectra. Unless otherwise stated, chemical shifts are given in ppm with the solvent as internal standard.
  • Amine 8 (2S)-8-(4-methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2-amine,
  • Acyl chlorides Acyl chloride 1 : 4-(trifiuoromethoxy)benzoyl chloride,
  • Acyl chloride 9 4-ethoxybenzoyl chloride
  • Acyl chloride 10 2,5-bis(trifluoromethyl)benzoyl chloride
  • Acid 27 4- ⁇ [(trifluoromethyl)sulfonyl]oxy ⁇ benzoic acid
  • Acid 32 6-isopropoxynicotinic acid.
  • Triethylamine (3 mole equivalent) and 2-(lH-benzotriazole-l-yl)-l, 1,3,3- tetramethyluronium hexafiuorophosphate (1.5 mole equivalent) were added to a solution of the carboxylic acid (1 mole equivalent) in anhydrous acetonitrile and he reaction mixture was stirred for 10 min.
  • a solution of the amine (1 mole equivalent) in anhydrous acetonitrile was added and the reaction mixture was stirred at ambient temperature for 1 h. The volatiles were removed in vacuo and the crude residue was purified by preparative HPLC.
  • Gene(s) encoding the full-length protein of the voltage-gated sodium channel of interest are cloned and expressed under a suitable promoter in a suitable cell line, as well known in the art.
  • the so constructed stable cell lines are used in screening assays to identify suitable compounds active on voltage-gated sodium channels. Suitable screening assays are as follows.
  • Li + influx assay The cell line expressing the voltage-gated sodium channel of interest is plated in conventional 96 or 384 well tissue plates at a suitable cell density (for example 40000 cells/well in 96 well plate, or 20000 cells/well in 384 well plate). The cells are then repeatedly washed with a suitable Na free buffer using a suitable commercially available washer (for example EL-405 washer) until all tissue culture medium is removed from the wells.
  • a suitable Na-free buffer could have the composition (mM) Choline chloride 137, KCl 5.4, MgSO 4 0.81, CaCl 2 0.95, glucose 5.55 and HEPES 25 at pH 7.4, but may also have other suitable composition. After completion of all wash steps, cells are incubated in the suitable Na free buffer for 15 min.
  • a buffer rich in LiCl for 60 min at 37 0 C.
  • the LiCl buffer is also enriched in potassium ions, causing a depolarizing stimulus to the cells.
  • Such a buffer may have the composition (mM): LiCl 100, KCl 50, MgSO 4 0.81, CaCl 2 0.95, glucose 5.55 and HEPES 25 at pH 7.4, but may also have other suitable composition.
  • an effective concentration for example 100 ⁇ M
  • the voltage-gated sodium channel opener veratridine, or any other suitable voltage-gated sodium channel opener may be added to the medium to enhance signal detection.
  • an effective concentration for example 10 ⁇ g/ml
  • suitable scorpion venom may also be added to the medium to delay channel inactivation.
  • the assay can be complemented with compounds from a compound library. Compounds of interest are added to the Li-rich solution, one in each well. At the end of the incubation period cells are repeatedly washed with Na free buffer until all extracellular LiCl is removed. Cell lysis is obtained through incubation of cells with triton (1%) for 15 min, or any other suitable method. The resulting cell lysate is then introduced into an atomic absorption spectrophotometer, thus quantifying the amount of Li-influx during the procedure described above.
  • the described assay can be run with any atomic absorption spectrophotometer using plates of 96-well format, 384-well format, or any other conventional plate format.
  • the described assay can be applied to cell lines expressing any given one or more of the voltage-gated sodium channel alpha subunits, as well as any given combination of one of the voltage- gated alpha subunits with any one or more beta subunit.
  • the cell line of choice can be further hyperpolarised by expression of a suitable potassium leak ion channel, for example TREK-I, either by transient co-transfection or through establishment of a stable co-transfected cell line.
  • a suitable potassium leak ion channel for example TREK-I
  • the successful expression of a leak K current can be verified using traditional intracellular electrophysiology, either in whole cell patch-clamp, perforated patch-clamp or conventional two-electrode voltage- clamp.
  • a cell line of choice modified to successfully express a voltage-gated sodium channel of interest together with a suitable potassium leak ion channel transfected can then be used for screening using atomic absorptions spectrometry, as described above.
  • Electrophysiological recordings of sodium currents in cells stably expressing the voltage- gated sodium channel of interest confirms activity and provides a functional measure of the potency of compounds that specifically inhibit such channels.
  • Electrophysiological studies can be performed using automated patch-clamp electrophysiology platforms, like IonWorks HT, Ion Works Quattro, PatchXpress, QPatch or any other suitable platform.
  • the cell line expressing the voltage-gated sodium channel of interest is appropriately prepared as suggested by the manufacturer of the automated patch-clamp platforms. Suitable extracellular and intracellular buffer for such experiments are applied according to the instructions given by the manufacturer of the automated patch- clamp platforms.
  • Cells that express the voltage-gated sodium channel protein of interest are exposed to drugs through the pipetting system integrated in the platforms.
  • a suitable voltage stimulus protocol is used to activate the voltage-gated sodium channel proteins of interest through depolarisation from a defined holding potential.
  • Electrophysiological studies can also be performed using the whole cell configuration of the standard patch clamp technique.
  • cells that express the voltage-gated sodium channel protein of interest are exposed to the drugs by conventional microperfusion systems and a suitable voltage stimulus protocol is used to activate the voltage-gated sodium channels.

Landscapes

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

Abstract

The present invention relates to new compounds and pharmaceutically-acceptable salts thereof, and to new intermediates used in the preparation thereof, pharmaceutical compositions containing said compounds and to the use of said compounds in therapy.

Description

NOVEL β-PIPERAZINE-TETRAHYDRONAPHTALENE DERIVATIVES FOR
THE TREATMENT OF PAIN
Field of the Invention
The present invention relates to new compounds, to pharmaceutical composition containing said compounds and to the use of said compounds in therapy. The present invention further relates to processes for the preparation of said compounds and to new intermediates useful in the preparation thereof.
Background
Voltage-gated sodium channels are critical elements in the control of electrical excitability of various cell types, including muscle and neuronal cells. In muscle and neuronal cells voltage-gated sodium channels are mainly responsible for the rising phase of the action potential. Voltage-gated sodium channels are composed of a single alpha subunit and one or two beta subunits. There are 10 known alpha subunit proteins, of which nine are functional as an ion channel. The different alpha subunit proteins are herein referenced to as Navl.x, with x being an integer between 1 and 9. This labelling is in accordance with the conventions of the International Pharmacological Association (REF). Alpha subunits are large proteins of an approximate weight of 260 kDA (~ 2000 amino acids), and are functional as voltage-gated sodium channels as monomeric structures. Four beta subunits are known at present. Beta subunits are smaller proteins of an approximate weight of 33-36 kDa. Beta subunits can modulate functional expression, as well as the characteristics of channel opening and closing (gating) of alpha subunits.
Five major lines of evidence support the notion that voltage-gated sodium channels are important therapeutic targets: a) the biophysical characteristics of voltage-gated sodium channels, b) the tissue expression pattern of voltage-gated sodium channels, c) evidence from preclinical research, d) the association between several congenital diseases and channelopathies of voltage-gated sodium channels, and e) evidence from the usage of pharmacological agents active at voltage-gated sodium channels in the clinic.
A main biophysical characteristic of voltage-gated sodium channels is the fast opening and closing (activation and inactivation) of the channel upon an appropriate voltage stimulus.
These features make voltage-gated sodium channels absolutely essential in the generation of the upstroke of the action potential in most neuronal and muscle cells, and thereby central to the functionality of such tissue. Thus, inhibitory pharmacological interference with the activity of NaVs is expected to have dampening effects on excitability of such tissue. Such agents may thus be useful in the treatment of diseases that involve hyperactivity of neuronal or muscle tissue.
As outlined above, there are nine functional alpha subunits of voltage-gated sodium channels. Each of these alpha subunits has a characteristic tissue expression pattern. Tissue-specific up- or down-regulation of the expression of several of the voltage-gated sodium channels in human diseases or preclinical disease models in animals strongly supports a central role for specific voltage-gated sodium channels in distinct diseases.
Navl.7 is expressed in human neuromas, which are swollen and hypersensitive nerves and nerve endings that are often present in chronic pain states (Acta Neurochirurgica (2002) 144(8) 803-810). Navl.7 is also expressed in dorsal root ganglion neurons and contributes to the small tetrodoxin (TTX) sensitive component seen in these cells. Navl.7 may thus be a potential pain target in addition to its role in neuroendocrine excitability (EMBO Journal (1995) 14(6) 1084-1090).
The present invention relates to a novel group of compounds that exhibit NaVl.7 inhibiting activity, and are therefore expected to be useful in the prophylaxis and treatment of different acute and chronic pain conditions.
WO 97/34883, WO 99/14212, WO 99/05135 and WO 99/14213 describe compounds for use in treatment of pain. The compounds described in these prior art documents bind to serotonine receptors. The compounds of the present invention have little to none activity towards the serotonine receptor. The compounds of the present invention also are contemplated to have an improved pharmacokinetic profile compared to the compounds in the prior art, including a higher oral bioavailability, a decreased clearance and a decreased volume of distribution. Without being bound to any theory, the difference in pharmacokinetic profile is believed to be due to the fact that the right hand side of the molecule is aromatic in the compounds of the present invention while this is not the case for the known compounds.
Disclosure of the Invention According to the invention there is provided compounds of formula I,
Figure imgf000004_0001
wherein:
Ri is phenyl or a heteroaromatic ring containing one or two heteroatoms selected from N, O and S and which may be independently mono- or di-substituted with R2 and/or R3, R2 and R3 are independently selected from Ci-C6 alkyl, halogen, CF3, OH, Ci-C3 alkoxy, OCF3, SO3CH3, SO3CF3, phenyl, phenyl-Ci-C6 alkyl and Ci-C6 alkyl phenyl; with the proviso that when Ri is phenyl with one substituent R2 which is Ci-C6 alkyl, halogen, or CF3, then Ri also has a second substituent R3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a pharmaceutically acceptable salt thereof.
In one embodiment of the invention there is provided compounds of formula I wherein Ri is pyrazolyl, thiophenyl or pyridinyl.
In another embodiment of the invention there is provided compounds of formula I wherein Ri is phenyl. In yet another embodiment of the invention there is provided compounds of formula I wherein Ri is phenyl, pyrazolyl, thiophenyl or pyridinyl which may be independently mono- or di-substituted with R2 and/or R3,
R2 and R3 are independently selected from Ci-C6 alkyl, halogen, CF3, OH, Ci-C3 alkoxy, OCF3, SO3CF3 and phenyl; with the proviso that when Ri is phenyl with one substituent R2 which is Ci-C6 alkyl, halogen, or CF3, then Ri also has a second substituent R3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention there is provided compounds of formula I wherein R2 and R3 are independently selected from Ci-C6 alkyl, halogen, CF3, Ci-C3 alkoxy, OCF3, SO3CF3, or phenyl.
In a further embodiment of the invention there is provided compounds of formula I wherein R2 and R3 are independently selected from CH3, C(CH3)3, F, Cl, CF3, OCH3, OCH2CH3, OCH2(CH3)2, OCF3, SO3CF3, or phenyl.
In one embodiment the invention there is provided compounds of formula Ia,
Figure imgf000005_0001
(Ia) wherein
Ri is phenyl or a heteroaromatic ring containing one or two heteroatoms selected from N, O and S and which may be mono- or di-substituted with R2 and/or R3, wherein R2 is Ci-C6 alkyl, halogen, CF3, OH, Ci-C3 alkoxy, OCF3, SO3CH3, SO3CF3, phenyl, phenyl-Ci-C6 alkyl, Ci-C6 alkyl phenyl, R3 is OH, CF3, OCF3, halogen, Ci-C6 alkyl or Ci-C3 alkoxy; with the proviso that when Ri is phenyl with one substituent R2 which is Ci-C6 alkyl, halogen, or CF3, then Ri also has a second substituent R3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a pharmaceutically acceptable salt thereof.
Definitions
For the avoidance of doubt it is to be understood that where in this specification a group is qualified by 'hereinbefore defined', 'defined hereinbefore' or 'defined above' the said group encompasses the first occurring and broadest definition as well as each and all of the other definitions for that group.
For the avoidance of doubt it is to be understood that in this specification Ci-C6 means a carbon group having 1, 2, 3, 4, 5 or 6 carbon atoms.
In this specification, unless stated otherwise, the term "alkyl" includes both straight and branched chain alkyl groups and may be, but are not limited to methyl, ethyl, n-propyl, i- propyl, n-butyl, i-butyl, s-butyl, t-butyl, n-pentyl, i-pentyl, neo-pentyl, n-hexyl or i-hexyl. The term having 1 to 4 carbon atoms and may be but are not limited to methyl, ethyl, n-propyl, i-propyl or t-butyl.
The term "alkoxy", unless stated otherwise, refers to radicals of the general formula -O-R, wherein R is selected from a hydrocarbon radical. The term "alkoxy" may include, but is not limited to methoxy, ethoxy, propoxy, isopropoxy, butoxy, t-butoxy or isobutoxy.
The term "amine" or "amino" refers to radicals of the general formula -NRR', wherein R and R' are independently selected from hydrogen or a hydrocarbyl radical. The term "aromatic" refers to hydrocarbyl radicals having one or more polyunsaturated carbon rings having aromatic character, (e.g., 4n + 2 delocalized electrons) and comprising 6 up to about 14 carbon atoms.
The term "aryl" used alone or as suffix or prefix, refers to a hydrocarbon radical having one or more polyunsaturated carbon rings having aromatic character, (e.g., 4n + 2 delocalized electrons) and comprising 5 up to about 14 carbon atoms, wherein the radical is located on a carbon of the aromatic ring.
The term 'alkylphenyl' refers to a phenyl group which is mono-, di-, or tri-substituted with alkyl such as for example methylphenyl in example 4.
The term "halo" or "halogen" refers to fluorine, chlorine, bromine and iodine radicals.
The term "heterocycle" or "heterocyclic" or "heterocyclic ring" refers to ring-containing monovalent and divalent radicals having one or more heteroatoms, independently selected from N, O, P and S, as part of the ring structure and comprising at least 3 and up to about 20 atoms in the rings preferably 5 and 6 membered rings. Heterocyclic ring may be saturated or unsaturated and contain one or more double bonds.
The term "heteroaryl" refers to heterocyclic monovalent and divalent radicals having aromatic character.
The term "heteroaromatic" refers to heterocyclic monovalent and divalent radicals having aromatic character and may be, but is not limited to oxazolyl, isoxazol, furyl, thiophenyl, pyrrolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, indolyl, indazolyl, pyridonyl or benzothienyl. Heteroaryl may also be quinolinyl, quinoxalinyl, naphthyridinyl, isoquinolinyl or thiazolyl.
The term "hydrocarbyl" refers to any structure comprising only carbon and hydrogen atoms up to 14 carbon atoms.
For the avoidance of doubt a group R3 substituted with a group SO3CF3, is an R3 substituted with trifiuoromenthanesulfonic acid.
It will be appreciated that throughout the specification, the number and nature of substituents on rings in the compounds of the invention will be selected so as to avoid sterically undesirable combinations. The term "mammal" includes any of various warm-blooded vertebrate animals of the class Mammalia, including but not limited to humans, generally characterized by a covering of hair on the skin.
Another embodiment of the invention relates to compounds selected from the group consisting of
5-Methyl-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-l- phenyl- 1 H-pyrazole-4-carboxamide,
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]biphenyl-3- carboxamide,
5-tert-Butyl-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yl]thiophene-2-carboxamide,
N-[(2S)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-4-
(trifluoromethoxy)benzamide, 4- { [(2S)-8-(4-Methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2-yl]carbamoyl}phenyl trifluoromethanesulfonate,
2,4-Dimethoxy-N-[(2R)-8-(4-methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2- yljbenzamide,
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-3,5- bis(trifluoromethyl)benzamide,
3-Fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-4-
(trifluoromethyl)benzamide,
6-Isopropoxy-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yl]nicotinamide, 2-Fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-5-
(trifluoromethyl)benzamide,
4-Ethoxy-N-[(2R)-8-(4-methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2- yljbenzamide,
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-2,5- bis(trifluoromethyl)benzamide,
2,4-Dichloro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yljbenzamide, 2-Fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-4- (trifluoromethyl)benzamide,
3-Fluoro-4-methyl-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yljbenzamide, 3-Chloro-4-fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yljbenzamide, and
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-6- (trifluoromethyl)nicotinamide, and pharmaceutically acceptable salts thereof.
For the avoidance of doubt the present invention relates to any one of the specific compounds mentioned above.
The present invention relates to the compounds of formula I or Ia as hereinbefore defined as well as to pharmaceutical acceptable salts thereof. Salts for use in pharmaceutical compositions will be pharmaceutically acceptable salts, but other salts may be useful in the production of the compounds of formula I or Ia.
A suitable pharmaceutically acceptable salt of the compounds of the invention is, for example, an acid-addition salt, for example a salt with an inorganic or organic acid. In addition, a suitable pharmaceutically acceptable salt of the compounds of the invention is an alkali metal salt, an alkaline earth metal salt or a salt with an organic base.
Other pharmaceutically acceptable salts and methods of preparing these salts may be found in, for example, Remington's Pharmaceutical Sciences (18th Edition, Mack Publishing
Co.). The compounds of the present invention may also exists as solvents, solvated hydrates or cocrystals.
The compounds of the invention may exhibit tautomerism. All tautomeric forms and mixtures thereof are included within the scope of the invention.
The compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation, or by derivatisation, for example with a homochiral acid followed by separation of the diastereomeric esters by conventional means (e.g. HPLC, chromatography over silica). All stereoisomers are included within the scope of the invention.
Compounds of the present invention have been named with the aid of computer software (ACDLabs 8.0 or 9.0/Name(IUPAC)).
Process
Another object of the invention relates to processes (a) or (b) for the preparation of compounds of general Formula I and salts thereof.
(a) acylation of a compound of formula II, with an acylating reagent such as a compound of formula III, wherein halo is fluoro, chloro or bromo and Ri is as defined in Formula I or
Ia.
Figure imgf000010_0001
The reaction may be carried out using a suitable acylating reagent such as an acyl chloride, in a suitable solvent such as dichloromethane, chloroform, toluene or acetonitrile at a temperature between -20 0C and reflux. The reaction is advantageously effected by the presence of a base. A suitable base may be an organic amine base such as pyridine, 2,6-lutidine, collidine, triethylamine, N,N-diisopropylethylamine, morpholine, 7V-methylmorpholine, diazabicyclo[5.4.0]undec-7-ene or tetramethylguanidine or an alkali metal or an alkaline earth metal carbonate or hydroxide such as sodium carbonate, potassium carbonate, calcium carbonate, sodium hydroxide or potassium hydroxide. The reaction may be aided by the presence of 4-dimethylaminopyridine.
(b) acylation of a compound of formula II, with a suitable carboxylic acid IV, wherein Ri is as defined in Formula I or Ia.
Figure imgf000011_0001
The transformation may be performed using a suitable activating reagent such as 2-(1H- benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate or N,N'- carbonyldiimidazole with a suitable base such as triethylamine or N ,N- diisopropylethylamine. The reaction may be performed in a suitable solvent such as dimethylformamide, acetonitrile or dichloromethane at a temperature between -20 0C and reflux.
Protection and deprotection of functional groups may take place before or after any of the reaction steps described hereinbefore.
Protecting groups may be removed in accordance with techniques which are well known to those skilled in the art and as described hereinafter.
The use of protecting groups is fully described in "Protective Groups in Organic Chemistry", edited by J.W.F. McOmie, Plenum Press (1973), and "Protective Groups in Organic Synthesis", 3rd edition, T.W. Greene & P.G.M. Wutz, Wiley-Interscience (1999).
Persons skilled in the art will appreciate that, in order to obtain compounds of the invention in an alternative, and, on some occasions, more convenient manner, the individual process steps mentioned herein may be performed in a different order, and/or the individual reactions may be performed at a different stage in the overall route (i.e. substituents may be added to and/or chemical transformations performed upon, different intermediates to those associated hereinbefore with a particular reaction). This will depend inter alia on factors such as the nature of other functional groups present in a particular substrate, the availability of key intermediates and the protecting group strategy (if any) to be adopted. Clearly, the type of chemistry involved will influence the choice of reagent that is used in the said synthetic steps, the need, and type, of protecting groups that are employed, and the sequence for accomplishing the synthesis.
It will also be appreciated by those skilled in the art that, although certain protected derivatives of compounds of formula I, which may be made prior to a final deprotection stage, may not possess pharmacological activity as such, they may be administered parenterally or orally and thereafter metabolised in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". Moreover, certain compounds of formula I may act as prodrugs of other compounds of formula I.
All prodrugs of compounds of formula I are included within the scope of the invention.
Medical use
The compounds of the invention exhibit voltage-gated sodium channel inhibiting activity, especially Navl .7 blocking activity, for example as demonstrated in the test described below. The present invention relates to the compounds of formula I or Ia, which inhibit any sodium channel. Modulation of voltage-gated sodium channels by pharmacological or genetical tools points to a central role for distinct voltage-gated sodium channels in several disease models. A mouse line has been generated which through advanced molecular biology technologies eliminates the functional expression of Navl.7 in DRG neurons that express Navl.8 (Proceedings of the National Academy of Sciences USA (2004) 101(34) 12706-12711). This mouse line shows greatly reduced pain responses in several pain behaviour models. Likewise, Herpes-vector mediated knockdown of Navl.7 in primary afferents of wildtype mice results in a decrease in inflammatory hyperalgesia (Human Gene Therapy (2005) 16(2) 271-277).
Antagonists of NaV channels have been shown to be useful for treating a variety of conditions, including acute and chronic nociceptive, visceral, inflammatory, central and peripheral neuropathic pain. More specifically, modulators of NaV activity are currently used or being tested in the clinic as anaesthetics, including local anaesthetics (Pain (2000) 87(1) 7-17), neuropathic pain reliefers (European Journal of Pain (2002) 6(Supplement 1) 61-68), acute pain reliefers (The Cochrane Database of Systematic Reviews (2005) 3), chronic pain reliefers (Pharmacotherapy (2001) 21(9) 1070-1081), inflammatory pain reliefers (Proceedings of the National Academy of Sciences USA (1999) 96(14) 7645- 7649), headache reliefers (Headache (2001) 41(Supplement 1) S25-S32). The compounds of the invention are thus expected to be useful in both the prophylaxis and the treatment of a condition which is effected or facilitated by inhibition of voltage-gated sodium channels, in particular pain, such as acute and chronic pain disorders including but not limited to widespread pain, localized pain, nociceptive pain, inflammatory pain, central pain, central and peripheral neuropathic pain, central and peripheral neurogenic pain, central and peripheral neuralgia, low back pain, postoperative pain, visceral pain, pelvic pain, allodynia, anesthesia dolorosa, causalgia, dysesthesia, fibromyalgia, hyperalgesia, hyperesthesia, hyperpathia, ischemic pain, sciatic pain, pain associated with cystitis, including but not limited to interstitial cystitis, pain associated with multiple sclerosis, pain associated with arthritis, pain associated with osteoarthritis, pain associated with rheumatoid arthritis and pain associated with cancer.
Other indications that may be treated with the compounds of the inventions are, but not limited to, migraine, pruritus, fibromyalgia, tinnitus and epilepsy.
The compounds of the present invention may be administered alone or in combination with other compounds, especially therapeutically active compounds.
The compounds of the present invention may for example be combined with one or more of the following therapeutically active compounds: proton pump inhibitors such as omeprazole, lansoprazole, rabeprazole, tentorpazole, pantoprazole, esomeprazole, revaprazan or sorprazan. Thus one embodiment of the invention relates to a combination wherein a compound of formula (I) or (Ia) or a pharmaceutically acceptable salt, solvate or in vivo hydrolysable ester, solvates, hydrated solvates, hydrates or co crystals thereof, or a pharmaceutical composition or formulation comprising a compound of formula (I) or (Ia) is administered concurrently, simultaneously, sequentially or separately with another pharmaceutically active compound or compounds selected from the following:
(i) antidepressants such as agomelatine, amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, pro trip tyline, ramelteon, reboxetine, robalzotan, sertraline, sibutramine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof, (ii) atypical antipsychotics including for example quetiapine and pharmaceutically active isomer(s) and metabolite(s) thereof. (iii) antipsychotics including for example amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutylpiperidine, pimozide, prochlorperazine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, ziprasidone and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(iv) anxiolytics including for example alnespirone, azapirones,benzodiazepines, barbiturates such as adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, quazepam, reclazepam, tracazolate, trepipam, temazepam, triazolam, uldazepam, zolazepam and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof. (v) anticonvulsants including for example carbamazepine, valproate, lamotrogine, gabapentin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof. (vi) Alzheimer's therapies including for example donepezil, memantine, tacrine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(vii) Parkinson's therapies including for example deprenyl, L-dopa, Requip, Mirapex,
MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(viii) migraine therapies including for example almotriptan, amantadine, bromocriptine, butalbital, cabergoline, dichloralphenazone, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pramipexole, rizatriptan, ropinirole, sumatriptan, zolmitriptan, zomitriptan, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(ix) stroke therapies including for example abciximab, activase, NXY-059, citicoline, crobenetine, desmoteplase,repinotan, traxoprodil and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof. (x) urinary incontinence therapies including for example darafenacin, falvoxate, oxybutynin, propiverine, robalzotan, solifenacin, tolterodine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(xi) neuropathic pain therapies including for example gabapentin, lidoderm, pregablin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof. (xii) nociceptive pain therapies such as celecoxib, etoricoxib, lumiracoxib, rofecoxib, valdecoxib, diclofenac, loxoprofen, naproxen, paracetamol and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(xiii) GABAb modulators such as baclofen, and equivalents and pharmaceutically active salts and metabolite(s) thereof. (xiv) Glutamate receptor antagonists and equivalents and pharmaceutically active salts and metabolite(s) thereof.
(xv) insomnia therapies including for example agomelatine, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, etomidate, glutethimide, halazepam, hydroxyzine, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, ramelteon, roletamide, triclofos,secobarbital, zaleplon, Zolpidem and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
(xvi) mood stabilizers including for example carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, verapamil, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
Such combination products employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active compound or compounds within therapeutically active dosage ranges and/or the dosage described in the publication reference.
One embodiment of the invention relates to compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, for use in therapy.
Another embodiment relates to the use of compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, in the manufacture of a medicament for treatment of pain. A further embodiment relates to the use of compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, in the manufacture of a medicament for treatment of acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain.
One embodiment relates to a method of treatment of pain, or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain in a patient suffering from, or at risk of, said disease, which comprises administering to the patient a therapeutically effective amount of a compounds of formula I or Ia as defined above, or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, or a pharmaceutical composition comprising said compounds. An agent for the treatment of pain, or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain, which comprises as active ingredient a compounds of formula I or Ia as defined above , or any of the specific compounds mentioned above, or pharmaceutically acceptable salts thereof, or any of the specific salts mentions for these compounds, or a pharmaceutical composition comprising said compounds.
Pharmaceutical compositions The compounds of the invention will normally be administered orally, subcutaneously, intravenously, intraarterially, transdermally, intranasally, by inhalation, or by any other parenteral route, in the form of pharmaceutical preparations comprising the active ingredient either as a free base or a non-toxic organic or inorganic acid addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions may be administered at varying doses.
One embodiment relates to a pharmaceutical composition comprising as active ingredient a therapeutically effective amount of the compound of formula I or Ia as defined above, in association with one or more pharmaceutically acceptable diluents, excipients and/or inert carriers.
Another embodiment relates to said pharmaceutical composition according, for use in the treatment of pain or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain.
Suitable daily doses of the compounds of the invention in therapeutic treatment of humans are about 0.005 to 25.0 mg/kg body weight at oral administration and about 0.005 to 10.0 mg/kg body weight at parenteral administration. Example of ranges of daily doses of the compounds of the invention in therapeutic treatment of humans are about 0.005 to 10.0 mg/kg body weight at oral administration and about 0.005 to 5.0 mg/kg body weight at parenteral administration. Compounds of the invention may also have the advantage that they may be more efficacious than, be less toxic than, have a broader range of activity than, be more potent than, be longer acting than, produce fewer side effects than, be more easily absorbed than, or that they may have other useful pharmacological properties over, compounds known in the prior art.
The invention is illustrated by way of the following examples.
Examples
General Methods
Starting materials used were available from commercial sources, or prepared according to literature procedures.
Microwave irradiation was performed in a Creator, Initiator or Smith Synthesizer Single -mode microwave cavity producing continuous irradiation at 2450 MHz.
Mass spectra were recorded on one of the following instruments: a Perkin-Elmer SciX API 150ex spectrometer; a VG Quattro II triple quadrupole; a VG Platform II single quadrupole; or a Micromass Platform LCZ single quadrupole mass spectrometer (the latter three instruments were equipped with a pneumatically assisted electrospray interface (LC- MS)).
Prep-HPLC: Preparative chromatography was run on Waters auto purification HPLC with a diode array detector. Column: XTerra MS C8, 19 x 300 mm, 10 μm. Gradient with acetonitrile/0.1 M ammonium acetate in 5 % acetonitrile in MiIIiQ Water, typically run from 20% to 60% acetonitrile, in 13 min. Flow rate: 20 mL/min. Alternatively, purification was achieved on a semi preparative Shimadzu LC-8A HPLC with a Shimadzu SPD-IOA UV- vis. -detector equipped with a Waters Symmetry® column (C 18, 5 μm, 100 mm x 19 mm). Gradient with acetonitrile/0.1 % trifiuoroacetic acid in MiIIiQ Water, typically run from 35% to 60% acetonitrile in 20 min. Flow rate: 10 mL/min. Alternatively, another column was used; Atlantis C18 19 x 100 mm, 5 μm column. Gradient with acetonitrile/0.1 M ammonium acetate in 5% acetonitrile in MiIIiQ Water, run from 0% to 35-50% acetonitrile, in 15 min. Flow rate: 15 mL/min. NMR spectra were recorded on a Varian Unity+ 400 NMR Spectrometer, operating at 400 MHz for proton and 100 MHz for carbon- 13, and equipped with a 5 mm BBO probe with Z-gradients; or on a Bruker av400 NMR spectrometer operating at 400 MHz for proton and 100 MHz for carbon-13, and equipped with a 3 mm flow injection SEI 1HZD-13C probehead with Z-gradients, using a BEST 215 liquid handler for sample injection; or on a Bruker DPX400 NMR spectrometer, operating at 400 MHz for proton and 100 MHz for carbon-13, and equipped with a 4-nucleus probe with Z-gradients. The following reference signals were used: TMS δ 0.00, or the residual solvent signal of DMSO-J6 δ 2.49, CD3OD δ 3.31 or CDCl3 δ 7.25 (unless otherwise indicated). Resonance multiplicities are denoted s, d, t, q, m and br for singlet, doublet, triplet, quartet, multiplet and broad, respectively. Column chromatography was performed using Merck Silica gel 60 (0.040-0.063 mm), or employing a Combi Flash® Companion system using RediSep normal-phase flash columns. Compounds have been named using ACD/Name, version 9.0, software from Advanced Chemistry Development, Inc. (ACD/Labs), Toronto ON, Canada, www.acdlabs.com, 2004.
Rotamers may or may not be denoted in spectra depending upon ease of interpretation of spectra. Unless otherwise stated, chemical shifts are given in ppm with the solvent as internal standard.
Reactants
Following reactants, either new (see synthetic procedures described below), commercially available or described in the literature, were used for the preparation of the target compounds (table 1):
Amines:
Amine 2: (2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-amine,
Amine 8 : (2S)-8-(4-methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2-amine,
Acyl chlorides: Acyl chloride 1 : 4-(trifiuoromethoxy)benzoyl chloride,
Acyl chloride 2: 3-fluoro-4-(trifluoromethyl)benzoyl chloride,
Acyl chloride 3: 2-fluoro-4-(trifluoromethyl)benzoyl chloride, Acyl chloride 6: 2,4-dimethoxybenzoyl chloride,
Acyl chloride 7: 3,5-bis(trifluoromethyl)benzoyl chloride,
Acyl chloride 8: 2-fluoro-5-(trifluoromethyl)benzoyl chloride,
Acyl chloride 9: 4-ethoxybenzoyl chloride, Acyl chloride 10: 2,5-bis(trifluoromethyl)benzoyl chloride,
Acyl chloride 11 : 2,4-dichlorobenzoyl chloride,
Acyl chloride 12: 3-fluoro-4-methylbenzoyl chloride,
Acyl chloride 13: 3-chloro-4-fluorobenzoyl chloride,
Acyl chloride 15: 6-(trifluoromethyl)nicotinoyl chloride, Car boxy Hc acids:
Acid 3: 5-methyl-l -phenyl- lH-pyrazole-4-carboxylic acid,
Acid 4: biphenyl-3-carboxylic acid,
Acid 5: 5-tøt-butylthiophene-2-carboxylic acid,
Acid 27: 4- {[(trifluoromethyl)sulfonyl]oxy} benzoic acid, Acid 32: 6-isopropoxynicotinic acid.
General methods of synthesis. Method A:
To a solution of the amine (1 mole equivalents) and triethylamine (1.2 mole equivalents) in anhydrous dichloromethane the acid chloride (1.2 mole equivalents) was added. The reaction mixture was stirred at ambient temperature for 5-30 min and concentrated in vacuo. The crude product was purified using preparative HPLC. Method B:
7V,7V'-Carbonyldiimidazole (1.2 mole equivalent) was added to a solution of the carboxylic acid (1.2 mole equivalent) in anhydrous dimethylformamide followed by addition of a solution of the amine (1 mole equivalent) in anhydrous dimethylformamide. The reaction mixture was heated at 80 0C for 2-12 h. Water was added and the mixture was extracted with ethyl acetate. The organic phase was dried over magnesium sulfate and concentrated in vacuo to yield the crude product. Purification by preparative HPLC. Method C:
Triethylamine (3 mole equivalent) and 2-(lH-benzotriazole-l-yl)-l, 1,3,3- tetramethyluronium hexafiuorophosphate (1.5 mole equivalent) were added to a solution of the carboxylic acid (1 mole equivalent) in anhydrous acetonitrile and he reaction mixture was stirred for 10 min. A solution of the amine (1 mole equivalent) in anhydrous acetonitrile was added and the reaction mixture was stirred at ambient temperature for 1 h. The volatiles were removed in vacuo and the crude residue was purified by preparative HPLC.
Table 1. The following compounds were prepared according to Methods A-C, using the reactants mentioned above: amines and counterparts which are either carboxylic acids or secondary amines.
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Biological tests
Expression of voltage-gated sodium channel in cell lines:
Gene(s) encoding the full-length protein of the voltage-gated sodium channel of interest are cloned and expressed under a suitable promoter in a suitable cell line, as well known in the art. The so constructed stable cell lines are used in screening assays to identify suitable compounds active on voltage-gated sodium channels. Suitable screening assays are as follows.
Li+ influx assay The cell line expressing the voltage-gated sodium channel of interest is plated in conventional 96 or 384 well tissue plates at a suitable cell density (for example 40000 cells/well in 96 well plate, or 20000 cells/well in 384 well plate). The cells are then repeatedly washed with a suitable Na free buffer using a suitable commercially available washer (for example EL-405 washer) until all tissue culture medium is removed from the wells. A suitable Na-free buffer could have the composition (mM) Choline chloride 137, KCl 5.4, MgSO4 0.81, CaCl2 0.95, glucose 5.55 and HEPES 25 at pH 7.4, but may also have other suitable composition. After completion of all wash steps, cells are incubated in the suitable Na free buffer for 15 min. Then, the Na free buffer is removed and cells are incubated with a buffer rich in LiCl for 60 min at 370C. The LiCl buffer is also enriched in potassium ions, causing a depolarizing stimulus to the cells. Such a buffer may have the composition (mM): LiCl 100, KCl 50, MgSO4 0.81, CaCl2 0.95, glucose 5.55 and HEPES 25 at pH 7.4, but may also have other suitable composition. To enhance signal-to-noise ratio, an effective concentration (for example 100 μM) of the voltage-gated sodium channel opener veratridine, or any other suitable voltage-gated sodium channel opener, may be added to the medium to enhance signal detection. Furthermore, and also to enhance signal-to-noise ratio, an effective concentration (for example 10 μg/ml) of suitable scorpion venom may also be added to the medium to delay channel inactivation. In order to find a modulator of the voltage-gated sodium channel of interest, the assay can be complemented with compounds from a compound library. Compounds of interest are added to the Li-rich solution, one in each well. At the end of the incubation period cells are repeatedly washed with Na free buffer until all extracellular LiCl is removed. Cell lysis is obtained through incubation of cells with triton (1%) for 15 min, or any other suitable method. The resulting cell lysate is then introduced into an atomic absorption spectrophotometer, thus quantifying the amount of Li-influx during the procedure described above.
The described assay can be run with any atomic absorption spectrophotometer using plates of 96-well format, 384-well format, or any other conventional plate format. The described assay can be applied to cell lines expressing any given one or more of the voltage-gated sodium channel alpha subunits, as well as any given combination of one of the voltage- gated alpha subunits with any one or more beta subunit.
If needed the cell line of choice can be further hyperpolarised by expression of a suitable potassium leak ion channel, for example TREK-I, either by transient co-transfection or through establishment of a stable co-transfected cell line. The successful expression of a leak K current can be verified using traditional intracellular electrophysiology, either in whole cell patch-clamp, perforated patch-clamp or conventional two-electrode voltage- clamp. A cell line of choice modified to successfully express a voltage-gated sodium channel of interest together with a suitable potassium leak ion channel transfected can then be used for screening using atomic absorptions spectrometry, as described above.
Whole-cell voltage clamp electrophysiology assay
Electrophysiological recordings of sodium currents in cells stably expressing the voltage- gated sodium channel of interest confirms activity and provides a functional measure of the potency of compounds that specifically inhibit such channels.
Electrophysiological studies can be performed using automated patch-clamp electrophysiology platforms, like IonWorks HT, Ion Works Quattro, PatchXpress, QPatch or any other suitable platform. The cell line expressing the voltage-gated sodium channel of interest is appropriately prepared as suggested by the manufacturer of the automated patch-clamp platforms. Suitable extracellular and intracellular buffer for such experiments are applied according to the instructions given by the manufacturer of the automated patch- clamp platforms. Cells that express the voltage-gated sodium channel protein of interest are exposed to drugs through the pipetting system integrated in the platforms. A suitable voltage stimulus protocol is used to activate the voltage-gated sodium channel proteins of interest through depolarisation from a defined holding potential.
Electrophysiological studies can also be performed using the whole cell configuration of the standard patch clamp technique. In this assay, cells that express the voltage-gated sodium channel protein of interest are exposed to the drugs by conventional microperfusion systems and a suitable voltage stimulus protocol is used to activate the voltage-gated sodium channels.
Example
Title compounds of the above Examples were tested in the Whole-cell voltage clamp electrophysiology assay described above and were found to exhibit IC50 values as shown in the table below.
Figure imgf000033_0001

Claims

1. A compounds of formula I,
Figure imgf000034_0001
wherein: Ri is phenyl or a heteroaromatic ring containing one or two heteroatoms selected from N, O and S and which may be independently mono- or di-substituted with R2 and/or R3, R2 and R3 are independently selected from Ci-C6 alkyl, halogen, CF3, OH, Ci-C3 alkoxy, OCF3, SO3CH3, SO3CF3, phenyl, phenyl-Ci-C6 alkyl and Ci-C6 alkyl phenyl; with the proviso that when Ri is phenyl with one substituent R2 which is Ci-C6 alkyl, halogen, or CF3, then Ri also has a second substituent R3, as (7?j-enantiomers, (Sj-enantiomers or a racemate in the form of a free base or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 wherein Ri is phenyl.
3. The compound according to any one of claims 1 to 2 wherein Ri is pyrazolyl, thiophenyl or pyridinyl.
4. The compound according to any one of claims 1 to 3 wherein R2 and R3 are independently selected from Ci-C6 alkyl, halogen, CF3, Ci-C3 alkoxy, OCF3, SO3CF3, or phenyl.
5. The compound according to any one of claims 1 to 3 wherein R2 and R3 are independently selected from CH3, C(CH3)3, F, Cl, CF3, OCH3, OCH2CH3, OCH2(CH3)2,
OCF3, SO3CF3, or phenyl.
6. Compounds selected from the group consisting of 5-Methyl-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-l- phenyl- 1 H-pyrazole-4-carboxamide,
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]biphenyl-3- carboxamide, 5-tert-Butyl-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yl]thiophene-2-carboxamide,
N-[(2S)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-4-
(trifluoromethoxy)benzamide,
4-{[(2S)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]carbamoyl}phenyl trifluoromethanesulfonate,
2,4-Dimethoxy-N-[(2R)-8-(4-methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2- yljbenzamide,
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-3,5- bis(trifluoromethyl)benzamide, 3-Fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-4-
(trifluoromethyl)benzamide,
6-Isopropoxy-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yl]nicotinamide,
2-Fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-5- (trifluoromethyl)benzamide,
4-Ethoxy-N-[(2R)-8-(4-methylpiperazin- 1 -yl)- 1 ,2,3 ,4-tetrahydronaphthalen-2- yljbenzamide,
N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-2,5- bis(trifluoromethyl)benzamide, 2,4-Dichloro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yljbenzamide,
2-Fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-4-
(trifluoromethyl)benzamide,
3-Fluoro-4-methyl-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yljbenzamide,
3-Chloro-4-fluoro-N-[(2R)-8-(4-methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2- yljbenzamide, and N-[(2R)-8-(4-Methylpiperazin-l-yl)-l,2,3,4-tetrahydronaphthalen-2-yl]-6-
(trifluoromethyl)nicotinamide, and pharmaceutically acceptable salts thereof.
7. The compound according to any one of claims 1 to 6 or a pharmaceutically-acceptable salts thereof, for use in therapy.
8. Use of the compound according to any one of claims 1 to 6 or a pharmaceutically- acceptable salts thereof, for the manufacture of a medicament for treatment of acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain.
9. A pharmaceutical composition comprising as active ingredient a therapeutically effective amount of the compound according to any one of claims 1 to 6 or a pharmaceutically-acceptable salts thereof, in association with one or more pharmaceutically acceptable diluents, excipients and/or inert carriers.
10. A method of treatment of pain disorders or acute or chronic nociceptive pain, visceral pain, inflammatory pain, and/or central or peripheral neuropathic pain, comprising administering to a mammal, including man in need of such treatment, a therapeutically effective amount of the compound according to any one of claims 1 to 6 or a pharmaceutically-acceptable salts thereof.
PCT/SE2008/050462 2007-04-23 2008-04-22 Novel 8-piperazine-tetrahydronaphtalene derivatives for the treatment of pain WO2008130323A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91331807P 2007-04-23 2007-04-23
US60/913,318 2007-04-23

Publications (1)

Publication Number Publication Date
WO2008130323A1 true WO2008130323A1 (en) 2008-10-30

Family

ID=39875743

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2008/050462 WO2008130323A1 (en) 2007-04-23 2008-04-22 Novel 8-piperazine-tetrahydronaphtalene derivatives for the treatment of pain

Country Status (1)

Country Link
WO (1) WO2008130323A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106632146A (en) * 2015-10-28 2017-05-10 南京圣和药业股份有限公司 A diynyl compound and a synthetic method thereof
WO2020054657A1 (en) 2018-09-10 2020-03-19 科研製薬株式会社 Novel heteroaromatic amide derivative and medicine containing same

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997034883A1 (en) * 1996-03-22 1997-09-25 Astra Aktiebolag Substituted 1,2,3,4-tetrahydronaphthalene derivatives
WO1999005134A1 (en) * 1997-07-25 1999-02-04 Astra Aktiebolag Substituted 1,2,3,4-tetrahydronaphthalene derivatives
WO1999014212A1 (en) * 1997-09-18 1999-03-25 Astrazeneca Ab Substituted chroman derivatives
WO1999014213A1 (en) * 1997-09-18 1999-03-25 Astrazeneca Ab Substituted chroman derivatives
WO2005013914A2 (en) * 2003-08-08 2005-02-17 Vertex Pharmaceuticals Incorporated Heteroarylaminosulfonylphenyl derivatives for use as sodium or calcium channel blockers in the treatment of pain
US20060025415A1 (en) * 2003-08-08 2006-02-02 Gonzalez Jesus E Iii Compositions useful as inhibitors of voltage-gated sodium channels

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997034883A1 (en) * 1996-03-22 1997-09-25 Astra Aktiebolag Substituted 1,2,3,4-tetrahydronaphthalene derivatives
WO1999005134A1 (en) * 1997-07-25 1999-02-04 Astra Aktiebolag Substituted 1,2,3,4-tetrahydronaphthalene derivatives
WO1999014212A1 (en) * 1997-09-18 1999-03-25 Astrazeneca Ab Substituted chroman derivatives
WO1999014213A1 (en) * 1997-09-18 1999-03-25 Astrazeneca Ab Substituted chroman derivatives
WO2005013914A2 (en) * 2003-08-08 2005-02-17 Vertex Pharmaceuticals Incorporated Heteroarylaminosulfonylphenyl derivatives for use as sodium or calcium channel blockers in the treatment of pain
US20050137190A1 (en) * 2003-08-08 2005-06-23 Gonzalez Jesus E.Iii Compositions useful as inhibitors of voltage-gated sodium channels
US20060025415A1 (en) * 2003-08-08 2006-02-02 Gonzalez Jesus E Iii Compositions useful as inhibitors of voltage-gated sodium channels

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106632146A (en) * 2015-10-28 2017-05-10 南京圣和药业股份有限公司 A diynyl compound and a synthetic method thereof
WO2020054657A1 (en) 2018-09-10 2020-03-19 科研製薬株式会社 Novel heteroaromatic amide derivative and medicine containing same
KR20210057008A (en) 2018-09-10 2021-05-20 가껭세이야꾸가부시기가이샤 Novel heteroaromatic amide derivatives and drugs containing the same

Similar Documents

Publication Publication Date Title
AU2008241610B2 (en) Novel n- (8-heteroaryltetrahydronaphtalene-2yl) or n- (5- heteroarylchromane-3-yl) carboxamide derivatives for the treatment of pain
JP3927630B2 (en) Preventive and therapeutic agents for viral infections
TWI477499B (en) Metabotropic glutamate receptor isoxazole ligands and their use as potentiators-286
US20040077697A1 (en) 2-Acylaminothiazole derivative or its salt
EP1797062B1 (en) Alkynyl based dervatives of benzophenone as non-nucleoside reverse transcriptase inhibitors
WO2000068203A1 (en) Cyclic compounds and uses thereof
EP1960347A2 (en) 17-beta-hydroxysteroid dehydrogenase type 3 (17beta-hsd3) inhibitors
KR20010034377A (en) Potassium Channel Inhibitors
JP2000128782A (en) Anilide derivative-containing ccr5 antagonist
KR102642063B1 (en) calcium channel inhibitor
KR20120088755A (en) Mglur4 allosteric potentiators, compositions, and methods of treating neurological dysfunction
JP2010502617A (en) Hydantoin derivatives useful as antibacterial substances
CA2611153A1 (en) 3-fluoro-piperidine t-type calcium channel antagonists
CN107814798B (en) 3-substituted acrylic acid compound and preparation method and application thereof
WO2001021206A1 (en) PREVENTIVES OR REMEDIES FOR MYOCARDITIS, DILATED CARDIOMYOPATHY AND CARDIAC INSUFFICIENCY CONTAINING NF-λB INHIBITORS AS THE ACTIVE INGREDIENT
KR102575605B1 (en) Manufacturing method of hepatitis B core protein modulator
WO2008130319A2 (en) Novel n-tetrahydronaphtalene or n-chromane carboxamide derivatives for the treatment of pain
WO2008130322A1 (en) Novel 5-heterocyclyl-chromane derivatives for the treatment of pain
WO2008130323A1 (en) Novel 8-piperazine-tetrahydronaphtalene derivatives for the treatment of pain
WO2008130321A2 (en) Novel n-tetrahydronaphtalene or 5-heterocyclyl-chromane or 8-heterocyclyl-tetrahydronaphtalene derivatives for the treatment of pain
KR20130095755A (en) Substituted 2-oxo- and 2-thioxo-dihydroquinoline-3-carboxamides as kcnq2/3 modulators
WO2011022315A1 (en) Pyrazinyl phenyl amide t-type calcium channel antagonists
EP3010900B1 (en) 1-sulfonyl piperidine derivatives as modulators of prokineticin receptors
EP2831071B1 (en) Imidazolyl methyl piperidine t-type calcium channel antagonists
WO2009105026A1 (en) Compound (r) -n*6*-ethyl-6, 7-dihydro-5h-indeno (5, 6- d) thiazole-2, 6-diamine and the use as antipsychotics

Legal Events

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

Ref document number: 08779263

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08779263

Country of ref document: EP

Kind code of ref document: A1