WO2007065449A1 - Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq - Google Patents

Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq Download PDF

Info

Publication number
WO2007065449A1
WO2007065449A1 PCT/DK2006/050039 DK2006050039W WO2007065449A1 WO 2007065449 A1 WO2007065449 A1 WO 2007065449A1 DK 2006050039 W DK2006050039 W DK 2006050039W WO 2007065449 A1 WO2007065449 A1 WO 2007065449A1
Authority
WO
WIPO (PCT)
Prior art keywords
disorder
alk
disorders
compounds
disease
Prior art date
Application number
PCT/DK2006/050039
Other languages
English (en)
Inventor
Nikolay Khanzhin
Daniel Rodriguez Greve
Mario Rottländer
Original Assignee
H. Lundbeck A/S
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 H. Lundbeck A/S filed Critical H. Lundbeck A/S
Priority to MX2008002294A priority Critical patent/MX2008002294A/es
Priority to CA002621854A priority patent/CA2621854A1/fr
Priority to BRPI0615631-2A priority patent/BRPI0615631A2/pt
Priority to EP06846967A priority patent/EP1937653A1/fr
Priority to JP2008529473A priority patent/JP2009507052A/ja
Priority to AU2006322461A priority patent/AU2006322461A1/en
Priority to EA200800780A priority patent/EA200800780A1/ru
Publication of WO2007065449A1 publication Critical patent/WO2007065449A1/fr
Priority to IL189545A priority patent/IL189545A0/en
Priority to NO20081713A priority patent/NO20081713L/no

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/50Three nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms

Definitions

  • the present invention relates to compounds, which are openers of the KCNQ family potassium ion channels.
  • the compounds are useful in the treatment of disorders and diseases being responsive to opening of the KCNQ family potassium ion channels, one such disease is epilepsy.
  • Ion channels are cellular proteins that regulate the flow of ions, including potassium, calcium, chloride and sodium into and out of cells. Such channels are present in all animal and human cells and affect a variety of processes including neuronal transmission, muscle contraction and cellular secretion.
  • KCNQ potassium channel genes
  • KCNQ4 gene is thought to encode the molecular correlate of a potassium channel found in outer hair cells of the cochlea and in Type I hair cells of the vestibular apparatus, in which mutations can lead to a form of inherited deafness.
  • KCNQl KvLQTl
  • KCNEl minimal K(+)-channel protein
  • Mutations in this channel can cause one form of inherited long QT syndrome type 1 (LQTl), as well as being associated with a form of deafness (Robbins Pharmacol Ther 2001, 90, 1-19).
  • KCNQ2 and KCNQ3 were discovered in 1988 and appear to be mutated in an inherited form of epilepsy known as benign familial neonatal convulsions (Rogawski Trends in Neurosciences 2000, 23, 393-398).
  • the proteins encoded by the KCNQ2 and KCNQ3 genes are localised in the pyramidal neurons of the human cortex and
  • hippocampus regions of the brain associated with seizure generation and propagation (Cooper et al. Proceedings National Academy of Science USA 2000, 97, 4914-4919).
  • KCNQ2 and KCNQ3 are two potassium channel subunits that form "M-currents" when expressed in vitro.
  • the M-current is a non- inactivating potassium current found in many neuronal cell types. In each cell type it is dominant in controlling membrane excitability by being the only sustained current in the range of action potential initiation (Marrion Annual Review Physiology 1997, 59, 483-504). Modulation of the M-current has dramatic effects on neuronal excitability, for example activation of the current will reduce neuronal excitability.
  • Openers of these KCNQ channels, or activators of the M-current, will reduce excessive neuronal activity and may thus be of use in the treatment of seizures and other diseases and disorders characterised by excessive neuronal activity, such as neuronal hyperexcitability including convulsive disorders, epilepsy and neuropathic pain.
  • Retigabine (D-23129; N-(2-amino-4-(4-fluorobenzylamino)-phenyl) carbamic acid ethyl ester) and analogues thereof are disclosed in EP554543.
  • Retigabine is an anti-convulsive compound with a broad spectrum and potent anticonvulsant properties, both in vitro and in vivo. It is active after oral and intraperitoneal administration in rats and mice in a range of anticonvulsant tests including: electrically induced seizures, seizures induced chemically by pentylenetetrazole, picrotoxin and N-methyl-D-aspartate (NMDA) and in a genetic animal model, the DBA/2 mouse (Rostock et al.
  • retigabine is active in the amygdala kindling model of complex partial seizures, further indicating that this compound has potential for anti-convulsive therapy. In clinical trials, retigabine has recently shown effectiveness in reducing the incidence of seizures in epileptic patients (Bialer et al. Epilepsy Research 2002, 51, 31-71).
  • KCNQ 2 and 3 channels have also been reported to be upregulated in models of neuropathic pain (Wickenden et al. Society for Neuroscience Abstracts 2002, 454.7), and potassium channel modulators have been hypothesised to be active in both neuropathic pain and epilepsy (Schroder et al. Neuropharmacology 2001, 40, 888-898).
  • KCNQ channel mRNA The localisation of KCNQ channel mRNA is reported in brain and other central nervous system areas associated with pain (Goldstein et al. Society for Neuroscience Abstracts 2003, 53.8). In addition to a role in neuropathic pain, the expression of mRNA for KCNQ 2-5 in the trigeminal and dorsal root ganglia and in the trigeminal nucleus caudalis implies that openers of these channels may also affect the sensory processing of migraine pain
  • KCNQ 2, 3 and 5 channels may help modulate synaptic activity in the CNS and contribute to the neuroprotective effects of KCNQ channel openers (Noda et al., Society for Neuroscience Abstracts 2003, 53.9). Retigabine and other KCNQ modulators may thus exhibit protection against the
  • KCNQ modulators may prevent neuronal damage induced by excessive neuronal activation, and such compounds may be of use in the treatment of neurodegenerative diseases, and be disease modifying (or antiepileptogenic) in patients with epilepsy.
  • anticonvulsant compounds such as benzodiazepines and chlormethiazole are used clinically in the treatment of the ethanol withdrawal syndrome and that other anticonvulsant compounds e.g. gabapentin are very effective in animal models of this syndrome (Watson et al. Neuropharmacology 1997, 36, 1369-1375), other anticonvulsant compounds such as KCNQ openers are thus expected to be effective in this condition.
  • mRNA for KCNQ 2 and 3 subunits are found in brain regions associated with anxiety and emotional behaviours such as bipolar disorder e.g. hippocampus and amygdala (Saganich et al.
  • KCNQ openers may be useful for the treatment of anxiety disorders and bipolar disorder.
  • WO 200196540 discloses the use of modulators of the M-current formed by expression of KCNQ2 and KCNQ3 genes for insomnia, while WO 2001092526 discloses that modulators of KCNQ5 can be utilized for the treatment of sleep disorders.
  • WO01/022953 describes the use of retigabine for prophylaxis and treatment of neuropathic pain such as allodynia, hyperalgesic pain, phantom pain, neuropathic pain related to diabetic neuropathy and neuropathic pain related to migraine.
  • WO02/049628 describes the use of retigabine for the treatment of anxiety disorders such as anxiety, generalized anxiety disorder, panic anxiety, obsessive compulsive disorder, social phobia, performance anxiety, post-traumatic stress disorder, acute stress reaction, adjustment disorders, hypochondriacal disorders, separation anxiety disorder, agoraphobia and specific phobias.
  • WO97/15300 describes the use of retigabine for the treatment of neurodegenerative disorders such as Alzheimer's disease; Huntington's chorea; sclerosis such as multiple sclerosis and amyotrophic lateral sclerosis; Creutzfeld- Jakob disease; Parkinson's disease; encephalopathies induced by AIDS or infection by rubella viruses, herpes viruses, borrelia and unknown pathogens; trauma- induced neurodegenerations; neuronal hyperexcitation states such as in medicament withdrawal or intoxication; and neurodegenerative diseases of the peripheral nervous system such as polyneuropathies and polyneuritides.
  • neurodegenerative disorders such as Alzheimer's disease; Huntington's chorea; sclerosis such as multiple sclerosis and amyotrophic lateral sclerosis; Creutzfeld- Jakob disease; Parkinson's disease; encephalopathies induced by AIDS or infection by rubella viruses, herpes viruses, borrelia and unknown pathogens; trauma- induced neurodegenerations; neuronal hyper
  • KCNQ channel openers have also been found to be effective in the treatment of stroke, therefore it can be expected that selective KCNQ openers are effective in the treatment of stroke (Schroder et al., Pflugers Arch., 2003; 446(5): 607-16; Cooper and Jan, Arch Neurol., 2003, 60(4):496-500; Jensen, CNS Drug Rev., 2002, 8(4):353-60).
  • KCNQ channels have been shown to be expressed in dopaminergic and cholinergic circuits in the brain that are associated with the brain's reward system, particularly the ventral tegmental area (Cooper et al., J Neurosci, 2001, 21, 9529-9540). Therefore, KCNQ channel openers are expected to be effective in hyperexcitability disorders that involve the brain's reward system such as cocaine abuse, nicotine withdrawal and ethanol withdrawal.
  • Potassium channels comprised of the KCNQ4 subunits are expressed in the inner ear (Kubisch et al., Cell., 1999 Feb 5;96(3):437-46) and opening of these channels is therefore expected to treat tinnitus.
  • novel compounds which are potent openers of the KCNQ family of potassium channels are also desired.
  • novel compounds with improved properties relative to known compounds, which are openers of the KCNQ family potassium channels, such as retigabine are also desired.
  • One object of the invention is the provision of compounds which are potent openers of the KCNQ family potassium channels.
  • the compounds of the invention are substituted pyrimidine derivatives of the below formula I or salts thereof
  • the invention provides a compound of formula I for use as a medicament.
  • the invention provides a pharmaceutical composition comprising a compound of formula I and a pharmaceutically acceptable carrier or diluent.
  • the invention provides the use of a compound of formula I for the preparation of a medicament for the treatment of seizure disorders, anxiety disorders, neuropathic pain and migraine pain disorders, other pain disorders, such as cancer pain, neurodegenerative disorders, stroke, cocaine abuse, nicotine withdrawal, ethanol withdrawal or hearing disorders, such as tinnitus.
  • the invention furthermore concerns the use of a compound of formula I in a method of treatment of seizure disorders, anxiety disorders, neuropathic pain and migraine pain disorders, other pain disorders, such as cancer pain, neurodegenerative disorders, stroke, cocaine abuse, nicotine withdrawal, ethanol withdrawal or hearing disorders, such as tinnitus.
  • heteroatom refers to a nitrogen, oxygen or sulphur atom.
  • Halogen means fluoro, chloro, bromo or iodo. "Halo” means halogen.
  • Amino designates NH 2 , which is attached to the remainder of the molecule via the nitrogen atom.
  • C 1-6 -alk(en/yn)yl means C 1-6 -alkyl, C 2-6 -alkenyl or C 2-6 -alkynyl.
  • C 1-6 -alkyl refers to a branched or unbranched alkyl group having from one to six carbon atoms, including but not limited to methyl, ethyl, prop-1-yl, prop-2-yl, 2-methyl- prop-1-yl, 2-methyl-prop-2-yl, 2,2-dimethyl-prop-l-yl, but-l-yl, but-2-yl, 3-methyl-but-l- yl, 3-methyl-but-2-yl, pent-1-yl, pent-2-yl, pent-3-yl, hex-l-yl, hex-2-yl and hex-3-yl.
  • C2 -6 -alkenyl refers to a branched or unbranched alkenyl group having from two to six carbon atoms and one double bond, including but not limited to ethenyl, propenyl and butenyl.
  • C2 -6 -alkynyl refers to a branched or unbranched alkynyl group having from two to six carbon atoms and one triple bond, including but not limited to ethynyl, propynyl and butynyl.
  • C 1-10 -alk(en/yn)yl means C 1-10 -alkyl, C 2-10 -alkenyl or C 2-10 -alkynyl.
  • C 1-10 -alkyl refers to a branched or unbranched alkyl group having from one to ten carbon atoms, including but not limited to methyl, ethyl, prop-1-yl, prop-2-yl, 2-methyl- prop-1-yl, 2-methyl-prop-2-yl, 2,2-dimethyl-prop-l-yl, but-l-yl, but-2-yl, 3-methyl-but- 1-yl, 3-methyl-but-2-yl, pent-1-yl, pent-2-yl, pent-3-yl, hex-l-yl, hex-2-yl, hex-3-yl, 2-methyl-4,4-dimethyl-pent- 1 -yl and hept- 1 -yl.
  • C2 -10 -alkenyl refers to a branched or unbranched alkenyl group having from two to ten carbon atoms and one double bond, including but not limited to ethenyl, propenyl and butenyl.
  • C2 -10 -alkynyl refers to a branched or unbranched alkynyl group having from two to ten carbon atoms and one triple bond, including but not limited to ethynyl, propynyl and butynyl.
  • C 3-8 -cycloalk(en)yl means C 3-8 -cycloalkyl or C 3-8 -cycloalkenyl.
  • C 3-8 -CyClOaIlCyI designates a monocyclic or bicyclic carbocycle having three to eight carbon atoms, including but not limited to cyclopropyl, cyclopentyl, cyclohexyl, bicycloheptyl such as 2-bicyclo[2.2.1]heptyl.
  • C 3-8 -cycloalkenyl designates a monocyclic or bicyclic carbocycle having three to eight carbon atoms and one double bond, including but not limited to cyclopentenyl and cyclohexenyl.
  • halo-C 1-6 -alk(en/yn)yl designates C 1-6 -alk(en/yn)yl being substituted with halogen, including but not limited to trifluoromethyl.
  • halo-C 1-6 -alk(en/yn)yloxy designates C 1-6 -alk(en/yn)yloxy being substituted with halogen, including but not limited to trifluoromethyloxy.
  • halo-C 3-8 -cycloalk(en)yl designates C 3-8 -cycloalk(en)yl being substituted with halogen, including but not limited to chlorocyclopropane and chlorocyclohexane.
  • halo-C 3-8 -cycloalk(en)yloxy designates C 3-8 -cycloalk(en)yloxy being substituted with halogen, including but not limited to chlorocyclopropyloxy and
  • halo-C 3-8 -cycloalk(en)yl-C 1-6 -alk(en/yn)yloxy designates halo-C 3-8 - cycloalk(en)yl being attached to the remainder of the molecule via C 1-6 -alk(en/yn)yloxy.
  • C 1-6 -alk(en/yn)yloxy designates C 1-6 -alk(en/yn)yl being attached to the remainder of the molecule via an oxygen atom.
  • C 3-8 -cycloalk(en)yloxy designates C 3-8 -cycloalk(en)yl being attached to the remainder of the molecule via an oxygen atom.
  • aryl designates monocyclic or bicyclic aromatic systems being selected from the group consisting of phenyl, naphthyl, thiophen, furan, benzothiophen and benzofuran.
  • optionally substituted aryl-C 1-6 -alk(en/yn)yl designates aryl-C 1-6 -alk(en/yn)yl wherein the aryl moiety is optionally substituted, such as with 1, 2 or 3 substituents independently selected from the group consisting of halogen, cyano, C 1-6 -alk(en/yn)yl, C 3-8 -cycloalk(en)yl, C 3-8 -cycloalk(en)yl-C 1-6 -alk(en/yn)yl, halo-C 1-6 -alk(en/yn)yl, halo- C 3-8 -cycloalk(en)yl, halo-C 3-8 -cycloalk(en
  • aryl designates aryl wherein the aryl is optionally substituted, such as with 1, 2 or 3 substituents independently selected from the group consisting of halogen, cyano, C 1-6 -alk(en/yn)yl, C 3-8 -cycloalk(en)yl, C 3-8 -cycloalk(en)yl- C 1-6 -alk(en/yn)yl, halo-C 1-6 -alk(en/yn)yl, halo-C 3-8 -cycloalk(en)yl, halo-C 3-8 -cycloalk(en)yl- C 1-6 -alk(en/yn)yl, C 1-6 -alk(en/yn)yloxy, C 3-8 -cycloalk(en)yloxy and C 3-8 -cycloalk(en)yl- C 1-6 -alk(en/yn)yloxy
  • the present invention relates to substituted pyrimidine derivatives which are potent openers of KCNQ potassium channels.
  • the present invention relates to a compound represented by the general formula I or salts thereof:
  • R 1 and R 2 are independently selected from the group consisting of hydrogen and optionally substituted aryl-C 1-6 -alk(en/yn)yl, provided that R 1 and R 2 are not both hydrogen, or R 1 and R 2 together with the nitrogen to which they are attached form a 5 to 7 membered ring optionally containing a further heteroatom;
  • R 3 and R 4 are independently selected from hydrogen, halogen, cyano, amino, C 1-6 - alk(en/yn)yl, C 3-8 -cycloalk(en)yl, halo-C 1-6 -alk(en/yn)yl, halo-C 3-8 -cycloalk(en)yl, C 1-6 -alk(en/yn)yloxy, C 3-8 -cycloalk(en)yloxy, C 3-8 -cycloalk(en)yl-C 1-6 -alk(en/yn)yloxy, halo-C 1-6 -alk(en/yn)yloxy, halo-C 3-8 -cycloalk(en)yloxy and halo-C 3-8 -cycloalk(en)yl- C 1-6 -alk(en/yn)yloxy, provided that R 3 and R 4 are not both hydrogen;
  • R 5 is selected from the group consisting of C 1-10 -alk(en/yn)yl, C 3-8 -cycloalk(en)yl-C 1-6 - alk(en/yn)yl, optionally substituted aryl-C 1-6 -alk(en/yn)yl and optionally substituted aryl; In one embodiment of the compound of formula I, q is 0. In another embodiment of the compound of formula I, q is 1.
  • R 1 and R 2 are independently selected from hydrogen and optionally substituted aryl-C 1-6 -alk(en/yn)yl, provided that R 1 and R 2 are not both hydrogen.
  • R 1 and R 2 together with the nitrogen to which they are attached form a 5 to 7 membered ring optionally containing a further hetero atom; in another embodiment said further hetero atom is oxygen; in another embodiment said ring is a 6 membered ring; in another embodiment said ring is a morpholine ring.
  • R 3 and R 4 are independently selected from amino and C 1-6 -alk(en/yn)yl, preferably methyl.
  • R 5 is selected from the group consisting of C 1-10 -alk(en/yn)yl, C 3-8 -cycloalk(en)yl-C 1-6 -alk(en/yn)yl, optionally substituted aryl-C 1-6 -alk(en/yn)yl and optionally substituted aryl.
  • a further embodiment concerns a compound of formula I as the free base or a salt thereof, said compound is selected from the compounds of the following scheme:
  • the present invention also comprises salts of the compounds of the invention, typically, pharmaceutically acceptable salts.
  • the salts of the invention include acid addition salts, metal salts, ammonium and alkylated ammonium salts.
  • the salts of the invention are preferably acid addition salts.
  • the acid addition salts of the invention are preferably pharmaceutically acceptable salts of the compounds of the invention formed with non-toxic acids.
  • Acid addition salts include salts of inorganic acids as well as organic acids. Examples of suitable inorganic acids include hydrochloric, hydrobromic, hydroiodic, phosphoric, sulfuric, sulfamic, nitric acids and the like.
  • suitable organic acids include formic, acetic, trichloroacetic, trifluoroacetic, propionic, benzoic, cinnamic, citric, fumaric, glycolic, itaconic, lactic, methanesulfonic, maleic, malic, malonic, mandelic, oxalic, picric, pyruvic, salicylic, succinic, methane sulfonic,
  • ethanesulfonic tartaric, ascorbic, pamoic, bismethylene salicylic, ethanedisulfonic, gluconic, citraconic, aspartic, stearic, palmitic, EDTA, glycolic, p-aminobenzoic, glutamic, benzenesulfonic, p-toluenesulfonic acids, theophylline acetic acids, as well as the
  • 8-halotheophyllines for example 8-bromotheophylline and the like.
  • pharmaceutical acceptable inorganic or organic acid addition salts include the
  • metal salts include lithium, sodium, potassium, magnesium salts and the like.
  • ammonium and alkylated ammonium salts include ammonium, methyl-, dimethyl-, trimethyl-, ethyl-, hydroxyethyl-, diethyl-, n-butyl-, sec-butyl-, tert-butyl-, tetramethylammonium salts and the like.
  • the compounds of this invention may exist in unsolvated as well as in solvated forms with pharmaceutically acceptable solvents such as water, ethanol and the like.
  • the solvated forms are considered equivalent to the unsolvated forms for the purposes of this invention.
  • the compounds of the present invention may have one or more asymmetric centre and it is intended that any optical isomers (i.e. enantiomers or diastereomers), as separated, pure or partially purified optical isomers and any mixtures thereof including racemic mixtures, i.e. a mixture of stereoisomers, are included within the scope of the invention.
  • any optical isomers i.e. enantiomers or diastereomers
  • pure or partially purified optical isomers i.e. a mixture of stereoisomers
  • Racemic forms can be resolved into the optical antipodes by known methods, for example, by separation of diastereomeric salts thereof with an optically active acid, and liberating the optically active amine compound by treatment with a base. Another method for resolving racemates into the optical antipodes is based upon chromatography on an optically active matrix. Racemic compounds of the present invention can also be resolved into their optical antipodes, e.g. by fractional crystallization. The compounds of the present invention may also be resolved by the formation of diastereomeric derivatives. Additional methods for the resolution of optical isomers, known to those skilled in the art, may be used. Such methods include those discussed by J. Jaques, A. Collet and S. Wilen in "Enantiomers, Racemates, and Resolutions", John Wiley and Sons, New York (1981). Optically active compounds can also be prepared from optically active starting materials or by stereoselective synthesis.
  • geometric isomers may be formed. It is intended that any geometric isomers, as separated, pure or partially purified geometric isomers or mixtures thereof are included within the scope of the invention. Likewise, molecules having a bond with restricted rotation may form geometric isomers. These are also intended to be included within the scope of the present invention. Furthermore, some of the compounds of the present invention may exist in different tautomeric forms and it is intended that any tautomeric forms that the compounds are able to form are included within the scope of the present invention. The invention also encompasses prodrugs of the present compounds, which on
  • prodrugs will be functional derivatives of the compounds of the general formula I, which are readily convertible in vivo into the required compound of the formula I.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
  • the invention also encompasses active metabolites of the present compounds.
  • the compounds according to the invention have affinity for the KCNQ2 receptor subtype with an EC 50 of less than 1500OnM such as less than 1000OnM as measured by the test "Relative efflux through the KCNQ2 channel” which is described below.
  • One embodiment concerns such compounds of formula I having affinity for the KCNQ2 receptor subtype with an EC 50 of less than 200OnM such as less than 150OnM as measured by the test "Relative efflux through the KCNQ2 channel” which is described below.
  • an embodiment concerns such compounds having affinity for the KCNQ2 receptor subtype with an EC 50 of less than 20OnM such as less than 15OnM as measured by the test "Relative efflux through the KCNQ2 channel" which is described below.
  • One embodiment concerns such compounds of formula I having an ED 50 of less than 15 mg/kg in the test "Maximum electroshock” which is described below. To further illustrate without limiting the invention an embodiment concerns such compounds having an ED 50 of less than 5 mg/kg in the test "Maximum electroshock” which is described below.
  • One embodiment concerns such compounds of formula I having an ED 50 of less than 5 mg/kg in the "Electrical seizure -threshold test” and “Chemical seizure -threshold test” which is described below.
  • One embodiment concerns such compounds of formula I having few or clinically insignificant side effects. Some of the compounds according to the invention are thus tested in models of the unwanted sedative, hypothermic and ataxic actions.
  • One embodiment concerns such compounds of formula I having a large therapeutic index between anticonvulsant efficacy and side-effects such as impairment of locomotor activity or ataxic effects as measured by performance on a rotating rod. Such compounds will expectedly be well tolerated in patients permitting high doses to be used before side effects are seen. Thereby compliance with the therapy will expectedly be good and administration of high doses may be permitted making the treatment more efficacious in patients who would otherwise have side effects with other medications.
  • the compounds according to the invention have effect on potassium channels of the KCNQ family, in particular the KCNQ2 subunit, and they are thus considered useful for increasing ion flow in a voltage-dependent potassium channel in a mammal such as a human.
  • the compounds of the invention are considered applicable in the treatment of a disorder or disease being responsive to an increased ion flow in a potassium channel such as the KCNQ family potassium ion channels.
  • Such disorder or disease is preferably a disorder or disease of the central nervous system.
  • the compounds of the invention may be administered as the only
  • the compounds of the invention may be administered as a part of a combination therapy, i.e. the compounds of the invention may be administered in combination with other therapeutically effective compounds having e.g. anti-convulsive properties.
  • other therapeutically effective compounds having e.g. anti-convulsive properties may include but not be limited to activities on:
  • ion channels such as sodium, potassium, or calcium channels
  • the excitatory amino acid systems e.g. blockade or modulation of NMDA receptors
  • the inhibitory neurotransmitter systems e.g. enhancement of GABA release, or blockade of GABA-uptake or
  • An aspect of the invention provides a compound of formula I or a salt thereof for use as a medicament.
  • the invention relates to the use of a compound of formula I or a salt thereof in a method of treatment.
  • An embodiment of the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I or a salt thereof and one or more pharmaceutically acceptable carrier or diluent.
  • the composition may comprise any of the embodiments of formula I as described above.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for increasing ion flow in a potassium channel of a mammal such as a human.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the treatment of a disorder or disease being responsive to an increased ion flow in a potassium channel, such disorder or disease is preferably a disorder or disease of the central nervous system.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of a disease or disorder wherein a KCNQ potassium channel opener such as a KCNQ2 potassium channel opener is beneficial.
  • such disorder or disease is selected from the group consisting of seizure disorders, anxiety disorders, neuropathic pain and migraine pain disorders, other pain disorders, such as cancer pain, neurodegenerative disorders, stroke, cocaine abuse, nicotine withdrawal, ethanol withdrawal or hearing disorders, such as tinnitus.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of seizure disorders.
  • the seizure disorders to be treated are selected from the group consisting of acute seizures, convulsions, status epilepticus and epilepsy such as epileptic syndromes and epileptic seizures.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of anxiety disorders.
  • the anxiety disorders to be treated are selected from the group consisting of anxiety and disorders and diseases related to panic attack, agoraphobia, panic disorder with agoraphobia, panic disorder without agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia and other specific phobias, obsessive-compulsive disorder, posttraumatic stress disorder, acute stress disorders, generalized anxiety disorder, anxiety disorder due to general medical condition, substance-induced anxiety disorder, separation anxiety disorder, adjustment disorders, performance anxiety, hypochondriacal disorders, anxiety disorder due to general medical condition and substance-induced anxiety disorder and anxiety disorder not otherwise specified.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of neuropathic pain and migraine pain disorders.
  • the neuropathic pain and migraine pain disorders to be treated are selected from the group consisting of allodynia, hyperalgesic pain, phantom pain, neuropathic pain related to diabetic neuropathy, neuropathic pain related to trigeminal neuralgia and neuropathic pain related to migraine.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of neurodegenerative disorders.
  • the neurodegenerative disorders to be treated are selected from the group consisting of Alzheimer's disease, Huntington's chorea, multiple sclerosis, amyotrophic lateral sclerosis, Creutzfeld- Jakob disease, Parkinson's disease, encephalopathies induced by AIDS or infection by rubella viruses, herpes viruses, borrelia and unknown pathogens, trauma- induced neurodegenerations, neuronal hyperexcitation states such as in medicament withdrawal or intoxication and neurodegenerative diseases of the peripheral nervous system such as polyneuropathies and polyneuritides.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of bipolar disorders or attention deficit hyperactivity disorder.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of sleep disorders; such as insomnia.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of fibromyalgia, a motor disorder or motion disorder, spasms, myokymia or urinary incontinence.
  • a further embodiment of the invention relates to the use of a compound of formula I or a salt thereof for the preparation of a pharmaceutical composition for the treatment of stroke, cocaine abuse, nicotine withdrawal, ethanol withdrawal or hearing disorders, such as tinnitus.
  • treatment as used herein in connection with a disease or disorders includes also prevention, inhibition and amelioration as the case may be.
  • the present invention also relates to a pharmaceutical composition.
  • the compounds of the invention or salts thereof may be administered alone or in combination with
  • compositions according to the invention may be formulated with
  • compositions may be specifically formulated for administration by any suitable route such as the oral, rectal, nasal, pulmonary, topical (including buccal and sublingual), transdermal, intracisternal, intraperitoneal, vaginal and parenteral (including subcutaneous, intramuscular, intrathecal, intravenous and intradermal) route, the oral route being preferred. It will be appreciated that the preferred route will depend on the general condition and age of the subject to be treated, the nature of the disorder or disease to be treated and the active ingredient chosen.
  • compositions formed by combining the compound of the invention and the pharmaceutical acceptable carriers are then readily administered in a variety of dosage forms suitable for the disclosed routes of administration.
  • the formulations may
  • the compounds of this invention are generally utilized as the free substance or as a pharmaceutically acceptable salt thereof.
  • One example is an acid addition salt of a compound having the utility of a free base.
  • a compound of the invention contains a free base such salts are prepared in a conventional manner by treating a solution or suspension of a free base of the invention with a chemical equivalent of a pharmaceutically acceptable acid. Representative examples are mentioned above.
  • compositions for oral administration may be solid or liquid.
  • Solid dosage forms for oral administration include e.g. capsules, tablets, dragees, pills, lozenges, powders, granules and tablette e.g. placed in a hard gelatine capsule in powder or pellet form or e.g. in the form of a troche or lozenge.
  • pharmaceutical compositions for oral administration may be prepared with coatings such as enteric coatings or they can be formulated so as to provide controlled release of the active ingredient such as sustained or prolonged release according to methods well known in the art.
  • Liquid dosage forms for oral administration include e.g. solutions, emulsions, suspensions, syrups and elixirs.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules or tablets, each containing a predetermined amount of the active ingredient, and which may include a suitable excipient.
  • the orally available formulations may be in the form of a powder or granules, a solution or suspension in an aqueous or non-aqueous liquid, or an oil- in- water or water-in-oil liquid emulsion.
  • Suitable pharmaceutical carriers include inert solid diluents or fillers, sterile aqueous solution and various organic solvents.
  • solid carriers are lactose, terra alba, sucrose, cyclodextrin, talc, gelatine, agar, pectin, acacia, magnesium stearate, stearic acid, lower alkyl ethers of cellulose, corn starch, potato starch, gums and the like.
  • liquid carriers are syrup, peanut oil, olive oil, phospho lipids, fatty acids, fatty acid amines, polyoxyethylene and water.
  • the carrier or diluent may include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax. Any adjuvants or additives usually used for such purposes such as colourings, flavourings, preservatives etc. may be used provided that they are compatible with the active ingredients.
  • the amount of solid carrier may vary but will usually be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation may be in the form of a syrup, emulsion, soft gelatine capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution.
  • Tablets may be prepared by mixing the active ingredient with ordinary adjuvants or diluents and subsequently compressing the mixture in a conventional tabletting machine.
  • compositions for parenteral administration include sterile aqueous and nonaqueous injectable solutions, dispersions, suspensions or emulsions as well as sterile powders to be reconstituted in sterile injectable solutions or dispersions prior to use. Depot injectable formulations are also contemplated as being within the scope of the present invention.
  • solutions of the compound of the invention in sterile aqueous solution aqueous propylene glycol, aqueous vitamin E or sesame or peanut oil may be employed.
  • aqueous solutions should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • the aqueous solutions are particularly suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • the sterile aqueous media employed are all readily available by standard techniques known to those skilled in the art.
  • Solutions for injections may be prepared by dissolving the active ingredient and possible additives in a part of the solvent for injection, preferably sterile water, adjusting the solution to the desired volume, sterilising the solution and filling it in suitable ampoules or vials.
  • Any suitable additive conventionally used in the art may be added, such as tonicity agents, preservatives, antioxidants, etc.
  • a typical oral dosage is in the range of from about 0.001 to about 100 mg/kg body weight per day, preferably from about 0.01 to about 50 mg/kg body weight per day, and more preferred from about 0.05 to about 10 mg/kg body weight per day administered in one or more dosages such as 1 to 3 dosages.
  • the exact dosage will depend upon the frequency and mode of administration, the sex, age, weight and general condition of the subject treated, the nature and severity of the disorder or disease treated and any concomitant diseases to be treated and other factors evident to those skilled in the art.
  • a typical unit dosage form for oral administration one or more times per day such as 1 to 3 times per day may contain from 0.01 to about 1000 mg, such as about 0.01 to 100 mg, preferably from about 0.05 to about 500 mg, and more preferred from about 0.5 mg to about 200 mg.
  • parenteral routes such as intravenous, intrathecal, intramuscular and similar
  • doses are in the order of about half the dose employed for oral administration.
  • a compound of the invention is meant any one of the embodiments of formula I as described herein.
  • the present invention relates to a method of preparing a compound of the invention as described in the following.
  • R 1 , R 2 , R 3 , R 4 , R 5 and q are as defined under formula I.
  • R 1 , R 2 , R 3 , R 4 , R 5 and q are as defined under formula I.
  • compounds which can exist as equilibrium between two or more tautomers only one tautomer is represented in the schemes, although it may not be to the most stable tautomer.
  • Such compounds include, but not limited to hydroxypyrimidines of the general formula IX, X, XVII, XVIII as well known to chemists skilled in the art.
  • Compounds of the general formula IV can be obtained by reacting compounds of the general formula II with amines of the general formula III with or without the addition of bases, such as trialkyl amines, potassium or sodium carbonate, in a suitable solvent, such as acetonitrile, iV,iV-dimethylformamide or ethanol, at a suitable temperature, such as room temperature, reflux temperature or at higher temperature under microwave irradiation in a sealed vessel.
  • bases such as trialkyl amines, potassium or sodium carbonate
  • a suitable solvent such as acetonitrile, iV,iV-dimethylformamide or ethanol
  • Compounds of the general formula V may be prepared from compounds of the general formula IV, by reducing the nitro group to an amino group, with suitable reducing agents such as zinc or iron powder in the presence of acid such as acetic acid or aqueous hydrochloric acid, or by hydrogen gas or ammonium formiate in the presence of a suitable hydrogenation catalyst such as palladium on activated carbon in suitable solvents such as methanol, ethanol, ethyl acetate or tetrahydroiuran, at suitable temperatures or under ultrasonic irradiation.
  • suitable reducing agents such as zinc or iron powder in the presence of acid such as acetic acid or aqueous hydrochloric acid, or by hydrogen gas or ammonium formiate in the presence of a suitable hydrogenation catalyst such as palladium on activated carbon in suitable solvents such as methanol, ethanol, ethyl acetate or tetrahydroiuran, at suitable temperatures or under ultrasonic irradiation.
  • Compounds of the invention of the general formula I may be prepared by reacting compounds of the general formula V with suitable electrophilic reagents, such as, but not limited to, suitably substituted carboxylic acid fluorides, carboxylic acid chlorides, carboxylic acid bromides, carboxylic acid iodides, carboxylic acid anhydrides, activated esters, chloroformates, and with or without the addition of bases, such as pyridine, trialkyl amines, potassium carbonate, magnesium oxide or lithium-, sodium-, or potassium alcoholates, in a suitable solvent, such as ethyl acetate, dioxane, tetrahydrofuran, acetonitrile or diethyl ether, at suitable temperatures, such as room temperature, reflux temperature or at higher temperature in a sealed vessel under microwave irradiation.
  • suitable electrophilic reagents such as, but not limited to, suitably substituted carboxylic acid fluorides, carboxylic acid chlorides, carboxylic acid brom
  • Activated esters and carboxylic acid anhydrides can be prepared from suitably substituted carboxylic acids under conditions known to chemists skilled in the art, for example as described by F. Albericio and L. A. Carpino, "Coupling reagents and activation” in Methods in enzymology: Solid- phase peptide synthesis, pp. 104-126, Academic Press, New York, 1997.
  • Carboxylic acid halides can be prepared from suitably substituted carboxylic acids by activation with reagents such as, but not limited to, thionyl chloride, oxalyl chloride, phosphorus tribromide or phosphorus triiodide under conditions well known to chemists skilled in the art.
  • Compounds of the general formula II may be prepared as outlined in Scheme 2.
  • Compounds of the general formula IX are prepared by condensation of urea with 1,3-dicarbonyl compounds VII or their equivalents such as unsaturated carbonyl compounds VIII in a suitable solvent such as iV,iV-dimethylformamide, JV-methylpyrrolidinone or ethanol, with or without addition of catalyst such as hydrochloric, sulphuric, methansulfonic or
  • Compounds of the general formula X may be prepared from compounds of the general formula IX 1 by nitration reactions known to chemists skilled in the art, such as reaction with concentrated nitric acid, sodium nitrite or sodium nitrate, in a suitable solvent, such as glacial acetic acid, acetic anhydride, trifluoroacetic acid, concentrated sulfuric acid or mixtures thereof, at appropriate temperatures, for example as described by P.B.D. de Ia Mare and J.H. Ridd, "Preparative methods of nitration" in Aromatic substitutions, pp.
  • Compounds of the general formula X may be converted into compounds of the general formula II by methods known to chemists skilled in the art such chlorination or bromination reaction with phosphorus oxychloride or phosphorus oxybromide.
  • Compounds of the general formula II, wherein X is fluorine or iodide can be prepared from the compounds of the general formula II, wherein X is chloride or bromide, by halogen exchange reaction with appropriate reagents such as hydroiodic acid, hydrofluoric acid, sodium iodide, potassiom fluoride under conditions known to chemists skilled in the art.
  • compounds of the general formula XX (XIII where R 4 is halogen) may be prepared from the compounds of the general formula XIX (XIII where R 4 is amino) by diazotization reaction followed by nucleophilic substitution in the presence of the appropriate halogen anion under conditions well known to chemists skilled in the art.
  • compounds of the general formula XIII 1 wherein R 4 is cyano may be prepared from compounds of the general formula XX (XIII where R 4 is halogen) by means of nickel-catalyzed cyanation reactions known to chemists skilled in the art for example as described by L. Cassar, J.Organomet.Chem., 1973, 54, C57-C58.
  • compounds of the general formula XIII wherein R 4 is C 1-6 -alk(en/yn)yloxy, C 3-8 -Cy cloalk(en)yloxy or C 3-8 -cycloalk(en)yl-C 1-6 -alk(en/yn)yloxy, may be prepared from compounds of general formula XX (XIII where R 4 is halogen) by reaction with the appropriate lithium-, sodium-, or potassium alcoholates or alcohols in the presence of base such as lithium-, sodium-, or potassium hydroxide, lithium-, sodium-, or potassium hydride, and with or without the addition of a catalyst such as copper sulfate, in a suitable solvent such as dioxane, at suitable temperatures, such as room temperature or reflux temperature.
  • base such as lithium-, sodium-, or potassium hydroxide, lithium-, sodium-, or potassium hydride
  • a catalyst such as copper sulfate
  • Microwave experiments were performed in sealed process vials or reactors using an Emrys Synthesizer or Emrys Optimizer EXP from Personal Chemistry or a Milestone Microsynth instrument from Milestone. When a reaction was heated in a microwave instrument, it was cooled to 25°C before the next process step.
  • Acid addition salts of the compounds of the invention may easily be formed by methods known to the person skilled in the art.
  • the assay measures the relative efflux through the KCNQ2 channel, and was carried out according to a method described by Tang et al. (Tang, W. et. al., J. Biomol. Screen. 2001, 6, 325-331) for hERG potassium channels with the modifications described below.
  • the compounds of the invention have an EC 50 of less than 2000OnM, in most cases less than 2000 nM and in many cases less than 200 nM. Accordingly, the compounds of the invention are considered to be useful in the treatment of diseases associated with the KCNQ family potassium channels.
  • Voltage-activated KCNQ2 currents were recorded from mammalian CHO cells by use of conventional patch-clamp recordings techniques in the whole-cell patch-clamp configuration (Hamill OP et.al. Pfl ⁇ gers Arch 1981; 391: 85-100).
  • CHO cells with stable expression of voltage-activated KCNQ2 channels were grown under normal cell culture conditions in CO 2 incubators and used for electrophysiological recordings 1-7 days after plating.
  • KCNQ2 potassium channels were activated by voltage steps up to + 80 mV in increments of 5-20 mV (or with a ramp protocol) from a membrane holding potential between - 100 mV and - 40 mV (Tatulian L et al. J Neuroscience 2001; 21 (15): 5535- 5545).
  • the electrophysiological effects induced by the compounds were evaluated on various parameters of the voltage-activated KCNQ2 current. Especially effects on the activation threshold for the current and on the maximum induced current were studied.
  • a left-ward shift of the activation threshold or an increase in the maximum induced potassium current is expected to decrease the activity in neuronal networks and thus make the compounds useful in diseases with increased neuronal activity - like epilepsia.
  • the test was conducted in groups of male mice using corneal electrodes and administering a square wave current of 26mA for 0.4 seconds in order to induce a convulsion characterised by a tonic hind limb extension (Wlaz et al. Epilepsy Research 1998, 30, 219-229).
  • Pilocarpine induced seizures are induced by intraperitoneal injection of pilocarpine 250mg/kg to groups of male mice and observing for seizure activity resulting in loss of posture within a period of 30 minutes (Starr et al. Pharmacology Biochemistry and Behavior 1993, 45, 321-325).
  • the threshold dose of pentylenetetrazol required to induce a clonic convulsion was measured by timed infusion of pentylenetetrazole (5mg / mL at 0.5 mL/minute) into a lateral tail vein of groups of male mice (Nutt et al. J Pharmacy and Pharmacology 1986, 38, 697-698).
  • the pharmacokinetic properties of the compounds were determined via. i.v. and p.o. dosing to Spraque Dawley rats, and, thereafter, drawing blood samples over 20 hours. Plasma concentrations were determined with LC/MS/MS.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Addiction (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Psychology (AREA)
  • Hospice & Palliative Care (AREA)
  • Anesthesiology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des dérivés de pyrimidine de formule générale I, ou des sels desdits dérivés, et leur emploi en tant qu'agents ouvrant les canaux ioniques potassiques de la famille KNCQ, en particulier dans le traitement de l'épilepsie.
PCT/DK2006/050039 2005-09-09 2006-09-07 Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq WO2007065449A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
MX2008002294A MX2008002294A (es) 2005-09-09 2006-09-07 Derivados de pirimidina sustituida.
CA002621854A CA2621854A1 (fr) 2005-09-09 2006-09-07 Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq
BRPI0615631-2A BRPI0615631A2 (pt) 2005-09-09 2006-09-07 composto, composição farmacêutica, e, uso de uma composição farmacêutica
EP06846967A EP1937653A1 (fr) 2005-09-09 2006-09-07 Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq
JP2008529473A JP2009507052A (ja) 2005-09-09 2006-09-07 ピリミジン誘導体およびそのkcnqカリウムチャネル開口薬としての使用
AU2006322461A AU2006322461A1 (en) 2005-09-09 2006-09-07 Pyrimidine derivatives and their use as KCNQ potassium channels openers
EA200800780A EA200800780A1 (ru) 2005-09-09 2006-09-07 Производные пиримидина и их применение в качестве открывателей калиевых каналов kcnq
IL189545A IL189545A0 (en) 2005-09-09 2008-02-14 Pyrimidine derivatives and their use as kcnq potassium channels openers
NO20081713A NO20081713L (no) 2005-09-09 2008-04-09 Pyrimidinderivater og deres anvendelse som KCNQ-kaliumkanalapnere

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200501262 2005-09-09
DKPA200501262 2005-09-09

Publications (1)

Publication Number Publication Date
WO2007065449A1 true WO2007065449A1 (fr) 2007-06-14

Family

ID=37199174

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2006/050039 WO2007065449A1 (fr) 2005-09-09 2006-09-07 Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq

Country Status (15)

Country Link
EP (1) EP1937653A1 (fr)
JP (1) JP2009507052A (fr)
KR (1) KR20080043314A (fr)
CN (1) CN101258133A (fr)
AR (1) AR055420A1 (fr)
AU (1) AU2006322461A1 (fr)
BR (1) BRPI0615631A2 (fr)
CA (1) CA2621854A1 (fr)
EA (1) EA200800780A1 (fr)
IL (1) IL189545A0 (fr)
MX (1) MX2008002294A (fr)
NO (1) NO20081713L (fr)
UA (1) UA93387C2 (fr)
WO (1) WO2007065449A1 (fr)
ZA (1) ZA200802174B (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009015667A1 (fr) * 2007-08-01 2009-02-05 H. Lundbeck A/S Utilisation d'agents d'ouverture kncq des canaux potassiques pour réduire des symptomes ou traiter des troubles et des états dans lesquels le système dopaminergique est détruit
WO2010097379A1 (fr) * 2009-02-24 2010-09-02 Neurosearch A/S Dérivés pyrimidine substitués et leur utilisation médicale
JP2010536771A (ja) * 2007-08-13 2010-12-02 バレアント ファーマシューティカルズ インターナショナル カリウムチャネルモジュレーターとしての5−アミノ−4,6−二置換インドールおよび5−アミノ−4,6−二置換インドリンの誘導体
WO2014000694A1 (fr) * 2012-06-29 2014-01-03 上海先声药物研究有限公司 Dérivé hétérocyclique de phendioxine
US10676438B2 (en) 2014-10-24 2020-06-09 Ono Pharmaceutical Co., Ltd. KCNQ2-5 channel activator

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2590961A1 (fr) * 2010-07-08 2013-05-15 Pfizer Inc Amides pipéridinyl pyrimidine en tant qu'ouvreurs du canal potassique ko7
MX367623B (es) * 2010-10-20 2019-08-29 Gruenenthal Gmbh 6-amino-nicotinamidas sustituidas como moduladores de kcnq2/3.
CN103508943B (zh) * 2012-06-29 2017-06-09 江苏先声药业有限公司 作为钾通道调节剂的化合物
US20220280455A1 (en) * 2019-08-02 2022-09-08 H. Lundbeck A/S Alcohol derivatives as kv7 potassium channel openers for use in epilepsy or seizures

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002066036A1 (fr) * 2001-02-20 2002-08-29 Bristol-Myers Squibb Company Derives de 2,4-disubstitue pyrimidine-5-carboxamide en tant que modulateur des canaux potassium kcnq
EP1334972A1 (fr) * 2002-02-12 2003-08-13 Pfizer Inc. Agents non peptidiques influant l'action de l'hormone de liberation de la gonadotrophine (GnRH)
WO2006064277A1 (fr) * 2004-12-17 2006-06-22 Takeda Cambridge Limited Derives de silicium et leur utilisation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002066036A1 (fr) * 2001-02-20 2002-08-29 Bristol-Myers Squibb Company Derives de 2,4-disubstitue pyrimidine-5-carboxamide en tant que modulateur des canaux potassium kcnq
EP1334972A1 (fr) * 2002-02-12 2003-08-13 Pfizer Inc. Agents non peptidiques influant l'action de l'hormone de liberation de la gonadotrophine (GnRH)
WO2006064277A1 (fr) * 2004-12-17 2006-06-22 Takeda Cambridge Limited Derives de silicium et leur utilisation

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BEBENBURG VON W ET AL: "Über substituierte Polyaminopyridine", CHEMIKER ZEITUNG, vol. 103, no. 12, 1979, pages 387 - 399, XP001117569 *
WICKENDEN ALAN D ET AL: "Retigabine, a novel anti-convulsant, enhances activation of KCNQ2/Q3 potassium channels", MOLECULAR PHARMACOLOGY, vol. 58, no. 3, September 2000 (2000-09-01), pages 591 - 600, XP000972243 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009015667A1 (fr) * 2007-08-01 2009-02-05 H. Lundbeck A/S Utilisation d'agents d'ouverture kncq des canaux potassiques pour réduire des symptomes ou traiter des troubles et des états dans lesquels le système dopaminergique est détruit
JP2011513196A (ja) * 2007-08-01 2011-04-28 ハー・ルンドベック・アクチエゼルスカベット ドーパミン作動系が破壊された障害もしくは状態の症状を軽減するためまたはその障害もしくは状態を処置するためのkcnqカリウムチャネル開口薬の使用
JP2010536771A (ja) * 2007-08-13 2010-12-02 バレアント ファーマシューティカルズ インターナショナル カリウムチャネルモジュレーターとしての5−アミノ−4,6−二置換インドールおよび5−アミノ−4,6−二置換インドリンの誘導体
WO2010097379A1 (fr) * 2009-02-24 2010-09-02 Neurosearch A/S Dérivés pyrimidine substitués et leur utilisation médicale
WO2014000694A1 (fr) * 2012-06-29 2014-01-03 上海先声药物研究有限公司 Dérivé hétérocyclique de phendioxine
US10676438B2 (en) 2014-10-24 2020-06-09 Ono Pharmaceutical Co., Ltd. KCNQ2-5 channel activator

Also Published As

Publication number Publication date
JP2009507052A (ja) 2009-02-19
UA93387C2 (ru) 2011-02-10
BRPI0615631A2 (pt) 2011-05-24
AU2006322461A1 (en) 2007-06-14
KR20080043314A (ko) 2008-05-16
ZA200802174B (en) 2009-10-28
CN101258133A (zh) 2008-09-03
CA2621854A1 (fr) 2007-06-14
IL189545A0 (en) 2008-06-05
AR055420A1 (es) 2007-08-22
MX2008002294A (es) 2008-03-14
EP1937653A1 (fr) 2008-07-02
EA200800780A1 (ru) 2008-06-30
NO20081713L (no) 2008-06-04

Similar Documents

Publication Publication Date Title
US7683058B2 (en) Substituted pyrimidine derivatives
CA2599890C (fr) Derives de pyridine substitues
US8012962B2 (en) Substituted thiomorpholine derivatives
US7601870B2 (en) Substituted aniline derivatives
WO2007065449A1 (fr) Derives de pyrimidine et leur emploi en tant qu'agents ouvrant les canaux potassiques kcnq
US7906537B2 (en) Substituted p-diaminobenzene derivatives
US20060264496A1 (en) Substituted indoline and indole derivatives
CA2523102A1 (fr) Derives indole et indoline substitues
IL185113A (en) Converted pyridine derivatives, pharmaceuticals containing it, and its use in drug preparation
MXPA06010329A (en) Substituted morpholine and thiomorpholine derivatives

Legal Events

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

Ref document number: 12008500331

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 565786

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 189545

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/002294

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020087004306

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200680032712.5

Country of ref document: CN

Ref document number: 2621854

Country of ref document: CA

Ref document number: 08024081

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2006322461

Country of ref document: AU

Ref document number: 2008529473

Country of ref document: JP

Ref document number: 1165/CHENP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008030408

Country of ref document: EG

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006846967

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2006322461

Country of ref document: AU

Date of ref document: 20060907

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006322461

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200800780

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 2006846967

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0615631

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080306