EP2753331A1 - Verwendung von 1h-chinazolin- 2,4 -dionen zur verwendung bei der prävention oder behandlung von lichtempfindlicher epilepsie - Google Patents

Verwendung von 1h-chinazolin- 2,4 -dionen zur verwendung bei der prävention oder behandlung von lichtempfindlicher epilepsie

Info

Publication number
EP2753331A1
EP2753331A1 EP11757513.4A EP11757513A EP2753331A1 EP 2753331 A1 EP2753331 A1 EP 2753331A1 EP 11757513 A EP11757513 A EP 11757513A EP 2753331 A1 EP2753331 A1 EP 2753331A1
Authority
EP
European Patent Office
Prior art keywords
methanesulfonamide
dioxo
dihydro
quinazolin
trifluoromethyl
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP11757513.4A
Other languages
English (en)
French (fr)
Inventor
Klaus KUCHER
Donald Johns
George IMBERT
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Original Assignee
Novartis AG
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 Novartis AG filed Critical Novartis AG
Publication of EP2753331A1 publication Critical patent/EP2753331A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/95Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in positions 2 and 4
    • C07D239/96Two oxygen 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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

Definitions

  • the present invention relates to pharmaceutical uses of 1H-quinazoline-2,4-diones, their pharmaceutically acceptable salts, and prodrugs thereof specifically for the treatment of photosensitive epilepsy (PSE).
  • PSE photosensitive epilepsy
  • Epilepsy is one of the most common neurological disorders, with lifetime prevalence in excess of 1 % of the world population. Despite the fact that there are about 20 antiepileptic drugs (AEDs) on the market, there is still a high medical need for improved treatments of epilepsy since about 30-40% of patients are inadequately controlled or suffer from drug side effects.
  • AEDs antiepileptic drugs
  • Photosensitive epilepsy is a rare form of reflex epilepsy in which seizures are triggered in photosensitive individuals by periodic visual stimuli such as flashing or flickering lights or regular patterns like stripes or checks.
  • the patterns are usually high in luminance contrast (bright flashes of light alternating with darkness, or white bars against a black background). Both natural and artificial light may trigger seizures.
  • Examples of flashing lights or rapidly changing or alternating images that may trigger the seizure in these patients include exposure to faulty lights or stroboscope lights such as those in disco-clubs, the light of emergency vehicles, images in films or television programs (with an increased risk of seizures with closer proximity to the light source); driving at dawn or dusk through an area in which the sun is shining through a line of trees or through a sudden change in light intensity (such as coming out of a tunnel); exposure to the light pattern caused by sun flickering on water; looking out from the window of a fast moving vehicle; or observing geometric patterns.
  • PSE is a type of reflex epilepsy and individuals with PSE may either have seizures exclusively in response to specific stimuli, not suffer spontaneous seizures or, alternatively, have reflex seizures coexisting with spontaneously occurring seizures.
  • PSE is mostly associated with generalized epilepsies.
  • the previously mentioned visual stimuli may provoke clinical photoconvulsive seizures or subclinical photoparoxysmal responses (PPR) in PSE patients.
  • Photosensitive epilepsy is a generalized epileptic-form reaction on intermittent photic stimulation (IPS) outlasting the stimulus sequence in about 5% of epileptic patients. Photosensitive epilepsy has a strong genetic component and a higher incidence in females.
  • the triggering stimuli vary from one patient to another, as do the nature and severity of the resulting seizures (ranging from brief absence to full tonic clonic seizure). Some patients are more sensitive with their eyes closed; others are more sensitive with their eyes open.
  • Ri is Ci-C 6 alkyl substituted by one, two or three substituents selected from hydroxy, d- C 6 alkoxy or C 5 -C 6 cycloalkoxy; or
  • R 3 is CrC 6 alkyl, hydroxy or (VCealkoxy-CrCealkyl; R 4 is hydrogen or CrC 6 alkyl; n is 1 or 2;
  • R 2 is CrC3alkyl or Ci-C 3 fluoroalkyl; or a pharmaceutically acceptable salt or prodrug thereof; for use in the treatment or prevention of photosensitive epilepsy.
  • a second aspect of the invention concerns a method for the treatment of photosensitive epilepsy in a subject in need of such treatment, which comprises administering to said subject a therapeutically effective amount of a 1 H-quinazoline-2,4-dione of formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
  • a third aspect of the invention relates to the use of a 1 H-quinazoline-2,4-dione of formula (I), or a pharmaceutically acceptable salt or prodrug thereof, for the treatment or prevention of photosensitive epilepsy.
  • a fourth aspect of the invention relates to a 1 H-quinazoline-2,4-dione of formula (I), or a pharmaceutically acceptable salt or prodrug thereof, for the treatment or prevention of photosensitive epilepsy.
  • a fifth aspect of the invention relates to a pharmaceutical composition comprising a 1 H- quinazoline-2,4-dione of formula (I), or a pharmaceutically acceptable salt or prodrug thereof, in the treatment or prevention of photosensitive epilepsy.
  • a sixth aspect of the invention relates to the use of a 1H-quinazoline-2,4-dione of formula (I), or a pharmaceutically acceptable salt or prodrug thereof, for the manufacture of a medicament for the treatment or prevention of photosensitive epilepsy.
  • a seventh aspect of the invention relates to a method for the treatment of photosensitive epilepsy in a subject in need of such treatment, which comprises administering to said subject a therapeutically effective amount of a 1 H-quinazoline-2,4-dione of formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
  • An eight aspect of the invention concerns a 1H-quinazoline-2,4-dione of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in combination with one or more antiepileptic drugs (AEDs), preferably one or two antiepileptic drugs (AEDs), for use in the treatment or prevention of photosensitive epilepsy.
  • AEDs antiepileptic drugs
  • AEDs antiepileptic drugs
  • a ninth aspect of the invention relates to a formulation comprising a compound of Formula (I) e.g. Compound C7, or a pharmaceutically acceptable salt or prodrug thereof , the compound having an AUC 2 4h greater than or equal to 5000 hr*ng/ml_ and/or C max greater than or equal to 300ng/ml e.g. such that the PPR is suppressed and/or the SPR is reduced, e.g. by at least 3 steps, optionally for use in the treatment of PSE
  • the invention relates to a 1 H-quinazoline-2,4-dione of formula (I) wherein
  • Ri is d-C 6 alkyl substituted by one, two or three substituents selected from hydroxy, d- C 6 alkoxy or C 5 -C 6 cycloalkoxy; C 5 -C 6 cycloalkyl substituted by one, two or three substituents selected from hydroxy, d-C 6 alkoxy or C 5 -C 6 cycloalkoxy; or
  • R 3 is C C 6 alkyl, hydroxy or Ci-Cealkoxy-Ci-Cealkyl;
  • R 4 is hydrogen or C C 6 alkyl;
  • n is 1 or 2;
  • R 2 is Ci-C 3 alkyl or d-Csfluoroalkyl; their pharmaceutically acceptable salts, and their prodrugs thereof; for use in a method for the treatment or prevention of photosensitive epilepsy.
  • the compound of formula (I) is a competitive AMPA antagonist. It is well understood that allosteric (non-competitive) antagonists provide an insurmountable blockade of AMPA receptors, potentially preventing any AMPA receptor-mediated neurotransmission at the synapse. In contrast, a high concentration of glutamate at the synapse can still activate the post-synaptic membrane in the presence of a competitive AMPA antagonist (albeit with a lower efficacy). Competitive AMPA antagonists may therefore exhibit an improved safety profile, as they will not fully block neurotransmission, but instead reduce the exaggerated glutamate signaling observed in some neurological disease, e.g. epilepsy.
  • Compounds of the formula (I) not only block AMPA-induced glutamate release from activated astrocytes but after oral dosing also suppress epilepsy seizures in epilepsy or in Rasmussen encephalitis.
  • the compound of the invention of formula (I) in addition to the advantage of being a competitive AMPA antagonist receptor inhibitor, presents also the advantage of being a selective competitive AMPA antagonist. Furthermore the compound of the invention of formula (I) is capable of penetrating the blood brain barrier and may be formulated in an oral dosage form.
  • Bonds with the asterisk (*) denote point of binding to the rest of the molecule.
  • CrC 6 alkyl represents a straight-chain or branched-chain alkyl group; for example, methyl, ethyl, n- or iso-propyl, n- iso- sec- or tert-butyl, n-pentyl, n-hexyl, with particular preference given to methyl, ethyl, n-propyl and iso-propyl.
  • C 5 -C 6 cycloalkyl represents cyclopentyl or cyclohexyl; preferably cyclopentyl.
  • alkyl/cycloalkyl-part of "alkoxy”, “cycloalkoxy”, “alkoxyalkyl” and “fluoroalkyl” shall have the same meaning as described in the above-mentioned definitions of "alkylTcycloalkyl".
  • CyCafluoroalkyl preferably represents trifluoromethyl, difluoromethyl orfluoromethyl.
  • any discussion of methods or references to the active ingredients includes said active ingredient in free form and in form of a pharmaceutically acceptable salt. If the active ingredients have, for example, at least one acidic center (for example COOH) they can form salts with bases.
  • the active ingredient or a pharmaceutically acceptable salt thereof may also be used in the form of a hydrate or may include other solvents used for crystallization.
  • a “pharmaceutically acceptable salt” is intended to mean a salt of a free base/free acid of a compound represented by formula (I) that is not toxic, biologically intolerable, or otherwise biologically undesirable.
  • Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of patients without undue toxicity, irritation, or allergic response.
  • Such salts are known in the field (e.g. S.M. Berge, et al, “Pharmaceutical Salts", J. Pharm. Sd., 1977, 66:1-19; and “Handbook of Pharmaceutical Salts, Properties, Selection, and Use", Stahl, RH., Wermuth, C.G., Eds.; Wiley-VCH and VHCA: Zurich, 2002).
  • the H-quinazoline-2,4-diones of formula (I) is used in free form.
  • the compounds may exist in optically active form or in form of mixtures of optical isomers, e.g. in form of racemic mixtures or diastereomeric mixtures. All optical isomers and their mixtures, including racemic mixtures, are part of the present invention.
  • the 1 H-quinazoline-2,4-dione of formula (I) is a compound, wherein R ! is Ci-C 6 alkyl substituted by one, two or three substituents selected from hydroxy, Ci-C 6 alkoxy or C 5 -C 6 cycloalkoxy; and R 2 is Ci-C 3 alkyl or CrCsfluoroalkyl.
  • the 1 H-quinazoline-2,4-dione of formula (I) is a compound, wherein is
  • R 3 is Ci-C 6 alkyl, hydroxy or CrCealkoxy-CrCealkyl; and R 2 is d-C 3 alkyl or Ci-C 3 fluoroalkyl.
  • the 1 H-quinazoline-2,4-dione of formula (I) is compound, wherein is
  • R 4 is hydrogen or CrC e alkyl; n is 1 or 2; and R 2 is d-C 3 alkyl or CrCafluoroalkyl.
  • the 1 H-quinazoline-2,4-dione of formula (I) is a compound selected from the group consisting of A-1 : N-[6-(1-Hydroxy-ethyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide; A-2: N-[6-(1 -Methoxy-ethyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-3 N-[6-(1 -Hydroxy-propy!)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-4 N-[6-(1-lsopropoxy-ethyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-5 N-[6-(1 -Ethoxy-ethyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-6 N-[2,4-Dioxo-6-(1 -propoxy-propyl)-7-trif !uoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-7 N-[6-(1 -isopropoxy-propyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-8 N-[7-Difluoromethyl-6-(1 -ethoxy-ethyl)-2,4-dioxo-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-9 N-[2,4-Dioxo-6-(1 -propoxy-ethyl)-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-10 N-[6-(1 -Butoxy-ethyl)-2,4-dioxo-7-trif luoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-11 N-[6-(1 -lsobutoxy-ethyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-12 N-[6-(1 -methoxy-butyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-13 N-[6-(1-Ethoxy-propyl)-2,4-dioxo-7-trifluoromethyl-1,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-14 N-[6-(1-Cyclopentyloxy-ethyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3- yl]-methanesulfonamide;
  • A-15 N-[6-(1 -Hydroxy-butyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-16 N-[6-(1 -Methoxy-2-methyl-propyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H- quinazolin-3-yl]-methanesulfonamide;
  • A-17 N-[6-(3-Hydroxy-propyl)-2,4-dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • A-18 N-[6-(1 -Hydroxy-3-methoxy-propyl)-2,4-dioxo-7-trif luoromethyl-1 ,4-dihydro-2H- quinazolin-3-yl]-methanesulfonamide;
  • B-2 N-[2,4-Dioxo-6-(tetrahydro-furan-2-yl)-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • B-3 N-[2 ⁇ -Dioxo-6-(tetrahydro-furan-3-yl)-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • C-1 N- ⁇ 7-lsopropyl-6-[2-(2-methoxy-ethyl)-2H-pyrazol-3-yl]-2,4-dioxo-1 ,4-dihydro-2H- quinazolin-3-yl ⁇ -met anesulfonamide;
  • C-5 N-[6-(2-Hydroxy-2H-pyrazol-3-yl)-2 ⁇ -dioxo-7-trifluoromethyl-1 ,4-dihydro-2H-quinazolin- 3-yl]-methanesulfonamide;
  • C-6 N-[7-Ethyl-6-(2-isopropyl-2H-pyrazol-3-yl)-2,4-dioxo-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • C-7 N-[7-lsopropyl-6-(2-methyl-2H-pyrazol-3-y!-2,4-dioxo-1 ,4-dihydro-2H-quinazo!in-3-yl]- methanesulfonamide;
  • C-8 N-[7-lsopropyl-6-(2-isopropyl-2H-pyrazol-3-yl)-2,4-dioxo-1 ,4-dihydro-2H-quinazolin-3-yl]- methanesulfonamide;
  • C-9 N-[7-Difluoromethyl-6-(2-methyl-2H-pyrazol-3-yl)-2,4-dioxo-1,4-dihydro-2H-quinazoli ⁇ yl]-methanesulfonamide
  • C-10 N-[7-Difluoromethyl-6-(2-isopropyl-2H-pyrazol-3-yl)-2,4-dioxo-1 ,4-dihydro-2H- quinazolin-3-yl]-methanesulfonamide;
  • C-14 N-[7-(1 -fluoro-ethyl)-6-(2-methyl-2H-pyrazol-3-yl)-2,4-dioxo-1 ,4-dihydro-2H-quinazolin- 3-yl]-methanesulfonamide;
  • C-15 N-[7-(1 , 1 -difluoro-ethyl)-6-(2-methyl-2H-pyrazol-3-yl)-2,4-dioxo-1 ,4-dihydro-2H- quinazolin-3-yl]-methanesulfonamide;
  • the compounds of the invention may be prepared by any suitable method, e.g. as described in WO 2006/108591.
  • the 1 H-quinazoline-2,4-dione of formula (I) is a compound selected from the group consisting of compound A-1 , A-2, A-3, A-4, A-5, A-6, A-7, A-8, A-9, A-10, A-11 , A-12, A- 3, A- 4, A-15, A- 6, A-17, A- 8 and A- 9. ln one embodiment of the invention, the 1 H-quinazoline-2,4-dione of formula (I) is a compound selected from the group consisting of compound B-1 , B-2 and B-3.
  • the 1 H-quinazoline-2,4-dione of formula (I) is a compound selected from the group consisting of compound C-1 , C-2, C-3, C-4, C-5, C-6, C- 7, C-8, C-9, C-10, C-11, C-12, C-13, C-14, C-15, C-16, C-17 and C-18.
  • Advantageous compounds of the invention i.e., the 1 H-quinazoline-2,4-diones of formula (I), should be well absorbed from the gastrointestinal tract, penetrate the blood brain barrier, be sufficiently metabolically stable and possess favorable pharmacokinetic properties.
  • Preferred compounds, having superior bioavailibility are 1 H-quinazoline-2,4-dione of formula (I) selected from the group consisting of compounds: A-1, A-2, A-3, A-4, A-5, A-6, A-7, A-13, A-14, A-15, A-18, B-2, B-3, C-1 , C-2, C-3, C-4, C-5, C-6, C-7, C-8, C-9, C-10, C-11 , C-12, C- 15, C-16, C-17 and C-18.
  • More preferred compounds, having superior bioavailibility are 1 H-quinazoline-2,4-dione of formula (I) selected from the group consisting of compounds: A-1 , A-2, A-3, A-4, A-5, A-7, A- 15, B-2, B-3, C-1 , C-2, C-3, C-6, C-7, C-8, C-9, C-10, C-11, C-12, C-15, C-17 and C-18.
  • 1H-quinazoline-2,4- dione of formula (I) selected from the group consisting of compounds: A-2, A-3, A-4, A-5 B-2, C-2, C-3, C-7, C-9, C-10, C- 1, C-15 and C-18.
  • 1 H-quinazoline-2,4-dione of formula (I) selected from the group consisting of compounds: A-2, A-5, B-2, C-7, C-9 and C- 11 , such as compound C-7.
  • Compounds for use in the present invention are either obtained in the free form, as a salt thereof, or as prodrug derivatives thereof.
  • prodrug as used herein relates to a compound, which converts in vivo into a compound used in the present invention.
  • a pro-drug is an active or inactive compound that is modified chemically through in vivo physiological action, such as hydrolysis, metabolism and the like, into a compound of this invention following administration of the prodrug to a subject.
  • the suitability and techniques involved in making and using pro-drugs are well known by those skilled in the art.
  • prodrug represents in particular compounds which are transformed in vivo to the parent compound, for example, by hydrolysis in blood, for example as described in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, Edward B.
  • Prodrugs therefore include drugs having a functional group which has been transformed into a reversible derivative thereof. Typically, such prodrugs are transformed to the active drug by hydrolysis. As examples may be mentioned the following:
  • Carboxylic acid Esters including e.g. alky I esters Alcohol Esters, including e.g. sulfates and phosphates as well
  • Amine Amides carbamates, imines, enamines,
  • Prodrugs also include compounds convertible to the active drug by an oxidative or reductive reaction. As examples may be mentioned
  • the compounds of the present invention can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
  • the compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms.
  • solvate refers to a molecular complex of a compound of the present invention (including pharmaceutically acceptable salts thereof) with one or more solvent molecules.
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like.
  • hydrate refers to the complex where the solvent molecule is water.
  • the compounds of the present invention including salts, hydrates and solvates thereof, may inherently or by design form polymorphs.
  • Preferred prodrugs of the invention should be well absorbed from the gastrointestinal tract, be transformed into the parent compound (or active principle, being the compound that in- vivo acts as AMPA receptor antagonist), the parent compound should be sufficiently metabolically stable and possess favorable pharmacokinetic properties.
  • prodrugs of the invention lead to an oral bioavailability of the parent compound which is comparable to the bioavailability when administered as a drug.
  • Further preferred prodrugs of the invention exhibit increased oral bioavailability compared to the parent compound when administered as a drug.
  • Oral bioavailability may manifest itself in different ways: (i) a biological effect may be achieved after oral administration when the parent compound is less effective upon oral administration, (ii) an earlier onset of action upon oral administration, (iii) a lower dose needed to achieve the same effect, (iv) a higher effect achieved by the same dose or (v) a prolonged action at the same dose.
  • Further preferred prodrugs of the invention are transformed into parent compounds which in- vivo bind potently to AMPA receptors whilst showing little affinity for other receptors.
  • Some prodrugs of the invention are transformed into parent compounds which also show antagonistic activity at kainate receptors. Besides such dual activity, showing little affinity for other receptors is a preferred feature. Further prodrugs of the invention - when the active principle is targeted against receptors in the central nervous system - are transformed into parent compounds that cross the blood brain barrier freely. Further prodrugs of the invention - when the active principle is targeted selectively against receptors in the peripheral nervous system - are transformed into parent compounds that do not cross the blood brain barrier.
  • Prodrugs, parent compounds and released pro-moieties should be non-toxic and demonstrate few side-effects.
  • the ideal prodrug of the invention will be able to exist in a physical form that is stable, non-hygroscopic and easily formulated.
  • the higher oral bioavailability of the compounds for use in the invention may give rise to the following beneficial effects relating to less bioavailable compounds: (i) an enhanced biological effect may be achieved after oral administration; (ii) an earlier onset of action may be observed following oral administration; (iii) a lower dose may be needed to achieve the same effect; (iv) a higher effect may be achieved by the same dose or (v) a prolonged action may be observed at the same dose.
  • the compound for use in the invention when tested in-vivo potently binds to AMPA receptors whilst showing little affinity for other receptors.
  • subject typically refers to a mammal, e.g. a human, especially to a human patient diagnosed with photosensitive epilepsy (PSE).
  • PSE photosensitive epilepsy
  • treatment refers to any type of treatment that imparts a benefit to a subject affected with photosensitive epilepsy, e.g. a human patient diagnosed with PSE, including prevention or reduction in number and severity of seizures.
  • terapéuticaally effective amount typically refers to a drug amount which, when administered to a subject, is sufficient to provide a therapeutic benefit, e.g. is sufficient for treating or preventing the photosensitive epileptic seizure (e.g. the amount provides an amelioration of symptoms, e.g. it leads to a reduction in number and severity of seizures).
  • the appropriate dosage will vary depending upon, for example, the compound employed, the host, the mode of administration and the nature and severity of the condition being treated. However, in general, satisfactory results in animals are indicated to be obtained at a daily dosage of from about 0.01 to about 100 mg/kg body weight, preferably from about 1 to about 30 mg/kg body weight, e.g. 10 mg/kg. In larger mammals, for example humans, an indicated daily dosage is in the range from about 0.1 to about 1000 mg, preferably from about 1 to about 400 mg, most preferably from about 10 to about 100 mg of a 1 H-quinazoline-2,4-dione of formula (I) conveniently administered, for example, in divided doses up to four times a day.
  • about 100mg of a 1 H-quinazoline-2,4-dione of formula (I) is administered daily. In a further embodiment, about 200mg of a 1 H-quinazoline-2,4-dione of formula (I) is administered daily.
  • the 1 H-quinazoline-2,4-diones of formula (I) may be administered as single active agent or in combination with one or more other active agents, in any usual manner, e.g. orally, for example in the form of tablets, capsules or drinking solutions; rectally, for example in the form of suppositories; intravenously, for example in the form of injection solutions or suspensions; or transdermally, for example in the form of a patch.
  • the manner of administration is oral administration, for example in the form of a tablet, capsule or drinking solution.
  • the manner of administration is rectal administration, for example in the form of a suppository.
  • the manner of administration is transdermal administration, for example in the form of a patch.
  • the manner of administration is oral administration.
  • Preferred pharmaceutical compositions comprise a 1 H-quinazoline-2,4-dione of formula (I) in association with at least one pharmaceutical carrier or diluent.
  • Such compositions may be manufactured in conventional manner.
  • Unit dosage forms may contain the compound of formula (I) e.g. compound C7 in an amount greater than or equal to 2.5mg, for example greater than or equal to 5mg, such as for example greater than or equal to 10mg or for example greater than or equal to 15mg.
  • Unit dosage forms may also contain the compound of formula (I) e.g. compound C7 in an amount of greater than or equal to 40mg, 50mg, 75mg or 100mg or greater than or equal to 150mg or 200mg.
  • Unit dosage forms may contain the compound of formula (I) e.g. compound C7 in an amount less than or equal to 300mg, for example less than or equal to 200mg, such as for example less than or equal to 150 mg or for example less than or equal to 100mg.
  • Unit dosage forms may also contain the compound of formula (I) e.g. compound C7 in an amount in the range from 5-200mg, e.g. 10-150mg or 15-100mg such as 50-1 OOmg.
  • compositions according to the invention are compositions for enteral administration, such as oral or rectal administration; or parenteral administration, such as intramuscular, intravenous, and nasal or transdermal administration, to warm-blooded animals (human beings and animals) that comprise an effective dose of the pharmacological active ingredient alone or together with a significant amount of a pharmaceutically acceptable carrier.
  • the dose of the active ingredient depends on the species of warm-blooded animal, body weight, age and individual condition, individual pharmacokinetic data, the disease to be treated and the mode of administration.
  • compositions comprise from approximately 1% to approximately 95%, preferably from approximately 20% to approximately 90%, active ingredient.
  • Pharmaceutical compositions according to the invention may be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, dragees, tablets or capsules.
  • compositions of the present invention may be prepared in a manner known per se, for example by means of conventional dissolving, lyophilizing, mixing, granulating or confectioning processes. Such processes are exemplified in WO W
  • the compound of formula (I) e.g. C7, which is comprised in the composition, may give an AUC 24 h or Cmax such that the PPR is suppressed and/or the SPR is reduced, e.g. by at least 3 steps.
  • the compound of formula (I) e.g. C7 which is comprised in the composition, may give an AUC 24 h greater than or equal to 5000 hr*ng/ml_, for example greater than or equal to 8000 hr * ng/mL, such as greater than or equal to 10000 hr * ng/ml_ or greater than or equal to 12000 hr * ng/mL or 15000 hr * ng/ml_.
  • C7 which is comprised in the composition, may also give an AUC 2 4h less than or equal to 25000 hr * ng/ml_, for example less than or equal to 22000 hr * ng/mL, such as less than or equal to 20000 hr*ng/mL.
  • the compound of formula (I) e.g. compound C7 which is comprised in the composition may give a C max of greater than or equal to 300ng/mL, such as greater than or equal to 400ng/mL or greater than or equal to 500ng/mL e.g greater than or equal to 750ng/mL, greater than or equal to 1000ng/ml_, greater than or equal to 400ng/mL, greater than or equal to 1800ng/mL or greater than or equal to 2400ng/mL such as greater than or equal to 2800ng/mL.
  • compound C7 which is comprised in the composition may also give a C max of less than or equal to 4000ng/mL, such as less than or equal to 3500ng/mL or less than or equal to 3000ng/mL. It will be understood that, for a given formulation, some variation in AUC 24 h or C max may be observed from patient to patient. The skilled person would understand that in this situation AUC 24h or Cmax is an aggregate value obtainable using a meaningful patient sample size e.g. as defined in EU or US clinical guidelines as in force at the filing date.
  • compositions for transdermal administration are described in Remington's Pharmaceutical Sciences 16 th Edition Mack; Sucker, Fuchs and Spieser, Pharmazeutician Technologie, 1 st Edition, Springer.
  • Epilepsy is usually diagnosed by observing seizures occurring spontaneously.
  • a seizure is a burst of abnormal electrical activity in the brain. Seizures can originate in a single location W
  • Photosensitive Epilepsy may be made by noting a correlation between the exposure to specific visual stimuli and seizure activity. This may be straightforward in situations where the seizures may impair the PSE subject's everyday life, e.g. by limiting his ability to drive a car. In other cases, patients suffering from PSE often are not recognized as affected by PSE as it is not always easy to identify that they are undergoing photosensitive epileptic episodes. Some seizures are so subtle that they can easily go unnoticed by the person experiencing them or by others. While the seizures themselves may not be noticed, the after-effects may include disabilities that linger for days: difficulties with mood, focus, memory, learning, sleep, sensory perception, and other functions.
  • Photosensitive epilepsy is a reflex form of epilepsy, in which epileptiform electroencephalogram (EEG) discharges may be evoked at any time by intermittent photic stimulation (IPS). This EEG response is also called photoparoxysmal response (PPR).
  • EEG epileptiform electroencephalogram
  • Each patient displays a different photosensitivity range, which is the difference between the lower and upper sensitivity limit to the visual stimulation. This range is related to liability of seizures in daily life.
  • the sensitivity range is specific for each patient and can be modified or abolished by using antiepileptic medicaments.
  • a standardized photoparoxysmal response range may be determined by performing the IPS with a standardized series of flash frequencies, for instance 14 frequencies from a range of 2 to 60 Hz.
  • SPR is a dimensionless parameter and is defined as the number of frequencies (steps) between the lowest and highest frequencies, e.g. between 2 and 60 Hz, that consistently elicit a PPR. If 14 frequencies are tested, the minimum and maximum possible SPR values are 0 and 14. Zero means complete abolition of reactivity on any stimulation frequency, i.e. complete abolishment of the PPR.
  • SPR is relatively stable for each patient and reflects the liability for seizures in daily life. The potential efficacy of an AED could be assessed by measuring hourly changes in the SPR after a single oral dose given to the PSE patients. The efficacy of the compounds of formula (I) in the treatment and prevention of PSE is confirmed by the following worked examples.
  • the SPR of each patient was the number of standard visual stimulation frequencies (in Hertz) that the patient was sensitive to, between the lower and upper thresholds. 14 frequencies were tested (from 2 to 60 Hertz).
  • the time of onset and the duration of response (PPR suppression or SPR reduction) to the treatment with Compound C-7 in patients with PSE and the maximal reduction in SPR in patients with PSE were also evaluated. Moreover in this study the pharmacokinetic profile of Compound C-7 in patients with PSE was measured.
  • BMI body mass index
  • SD standard deviation
  • n patients with non-missing data points
  • N analysis set total.
  • the selected patients had a documented diagnosis of epilepsy for at least 6 months prior to initial dosing of compound C-7.
  • the selected patients showed a consistent PPR during EEG evaluations prior to the first dosing indicating a diagnosis of photosensitive epilepsy.
  • Women of child-bearing potential (WOCBP) were asked to use acceptable methods of contraception.
  • Patients with an SPR value of 3 or less at the screening and patients with no consistent PPR (less than 3 SPR steps difference between the two evaluations required) when stimulated by intermittent photic stimulation (IPS), as determined by comparison between screening and pre-dose Day 1 were excluded from the study.
  • the dose of the compound of formula C-7 in Cohort studies I, II and III was 50, 100 and 15 mg respectively.
  • Some patients in Cohort I were treated with one concomitant AED from a pre-selected list of AEDs (valproate, lamotrigine, levetiracetam, clobazam, topiramate, pregabalin, gabapentin, zonisamide).
  • Some patients in Cohort II and III were treated with a combination of two concomitant AEDs from a preselected list of AEDs devoid of interaction potential with the compound of formula C-7 (valproate, lamotrigine, levetiracetam, clobazam, topiramate, zonisamide).
  • a stable dosing regimen was required for at least 4 weeks prior to initial dosing, and throughout the entire study.
  • AEDs Antiepileptic Drugs
  • the allowed AEDs were: valproate, lamotrigine, levetiracetam, clobazam, topiramate and zonisamide
  • the C-7 PK parameters were determined in plasma using non-compartmental methods using WinNonlin Pro (Version 5.2), as detailed in Table 3.
  • AUCiast The area under the concentration-time curve from time zero to the last measurable concentration sampling time [ng/mL.h]
  • Biofluid concentrations were expressed in mass per volume units (ng/mL). All C-7 and AEDs concentrations below LLOQ were reported as zero in the concentration data listings. Concentrations below LLOQ were treated as zero in summary statistics of concentration data as well as for calculation of C-7 PK parameters. A geometric mean was not reported whenever at least one concentration was below the LLOQ (i.e. zero). Missing data were labeled as such and no imputation methods were used.
  • Terminal elimination rate constant (lambda_z) was calculated as the slope of the linear regression of the terminal phase of the logarithmic concentration-time profile for each individual dataset available. A minimum of three time points was used for determining lambda_z. Regression was performed without weighting, and a minimum value of 0.75 for Rsq_adjusted was necessary for acceptance.
  • AED concentrations were listed and summarized; analysis of level of exposure of AED(s) taken by the patient was performed on an ad-hoc basis as it depended on specifics of the AED.
  • Compound C-7 15 mg dose group showed a numerically lower but still measurable PD effect compared to 50 and 100 mg dose groups.
  • C-7 100 mg dose group The majority of patients had response onset within 1 to 2 hours of dosing. Time to maximum reduction of SPR was shorter in C-7 100 mg dose group compared to the other 2 dose groups. The duration of response was similar in C-7 50 and 100 mg dose groups, ranging from 23 to 34 hours inclusive. Two patients in C-7 50 mg group and 1 patient in C-7 100 mg group maintained SPR response (i.e., stayed in responding status at all time points between first and last response), i.e. 33 hours in both patients. In the C-7 15 mg dose group, the response was noted to be intermittent in all eye conditions and none of the patients maintained the SPR response. Suppression was observed in 7 of the 13 patients, with the onset ranging from 1 to 4 hours after dosing and the duration ranging from 2 to 32 hours.
  • Table 5 summarizes the C-7 PK parameters after a single dose administration of 15, 50 and 100 mg to male and female patients with PSE.
  • C-7 was measurable in the plasma as early as 0.25 h post-dose following single administration of a 15, 50 or 100 mg dose to patients with PSE.
  • the plasma concentration of C-7 peaked at around 3 h post-dose (median) with minimum values of approximately 2h and maximum values of 4h in individual patients.
  • the pharmacokinetic data of C-7 in the patients with PSE in all 3 cohorts appeared to be overlapping with those in male healthy subjects observed in healthy volunteer study, although mean exposure in patients with PSE tended to be slightly higher.
  • a comparison of the means demonstrates that in patients mean C max was about 10-20% and mean AUC
  • T ag (hr) 1 0.00 (0.00-0.00) 0.00 (0.00-0.00) 0.00 (0.00-0.00)
  • a PK-PD model was developed sequentially for the SPR data. Firstly a pharmacokinetic model was fitted using nonlinear mixed effects methodology in NONMEM version VI to describe the 3 patients with both PK and PD data. A 3 compartment disposition model with first order absorption and absorption lag time was selected to describe the pharmacokinetic time profile for each patient. The between patient variability was described by exponential random effects on each pharmacokinetic parameter, with all parameters assumed to be independent between individuals. The residual error model combined the additive and proportional error terms. The estimated pharmacokinetic parameters for each patient were used to generate each patient's PK profile as an input to the pharmacodynamic model.
  • the mean (SD) EC 50 for eyes open, eyes closed and eyes closures was 2964 ng/ml (4235 ng/ml), 3746 ng/ml (4475 ng/ml), 2252 ng/ml (3505 ng/ml) respectively. This wide variability was the result of either many patients having either a low estimated EC 50 (5 ng/ml) or high estimated EC5 0 (9900 ng/ml).
  • the mean (SD) E max on the logit scale for eyes open, eyes closed and eyes closures was 21.6 (23.5), 16.3 (21.7), 12.9 (19.6) respectively suggesting there was a substantial reduction in SPR in the 3 eye conditions. Table 6 - Pharmacodynamic model parameter estimates for each patient Eyes open Eyes closed Eyes closure
  • Base Emax EC50 Base Emax ECso Base Emax EC50
  • the maximum and minimum AUC0-24h between which relevant reduction of SPR was noted to be maintained was 15334 hr*ng/ml_ (patient I; C-7 100 mg group) and 11624 hr*ng/ml_ (patient D; C-7 50 mg group), respectively
  • the C ma x between which suppression of PPR was noted to be maintained was 3700 ng/mL (patient H, C-7 100 mg group) and 1530 ng/mL (patient A; C-7 50 mg group), respectively; and the C max , between which relevant reduction of SPR was noted to be maintained was 2490 ng/mL (patient E, C-7 50 mg group) and 2070 ng/mL (patient I; C-7 100 mg group), respectively.
  • the maximum and minimum AUCo ⁇ between which suppression of PPR was noted to be maintained was 21302 hr*ng/ml_ (patient H; C-7 100 mg group) and 10056 hr*ng/mL (patient A; C-7 50 mg group), respectively.
  • the maximum and minimum AUC 0 -24h between which relevant reduction of SPR ( ⁇ 3 steps) was noted to be maintained was 15334 hr*ng/ml_ (patient I; C-7 100 mg group) and 11624 hr*ng/ml_ (patient D; C-7 50 mg group), respectively
  • the maximum and minimum C max between which suppression of PPR was noted to be maintained was 3700 ng/mL (patient H, C-7 100 mg group) and 530 ng/mL (patient A; C-7 50 mg group), respectively.
  • the maximum and minimum C max between which relevant reduction of SPR was noted to be maintained was 2490 ng/mL (patient E, C-7 50 mg group) and 2070 ng/mL (patient I; C-7 00 mg group), respectively.
  • Oral bioavailability of the compounds of the invention may be demonstrated using any generally known test in which the compound is administered orally and a biological effect observed.
  • Oral bioavailability of the compounds of the invention in the treatment of photosensitive epilepsy may be further quantified by the Maximal Electroshock test, which demonstrates that the compounds are orally bioavailable, penetrate the blood brain barrier and bind to the target receptor.
  • the oral bioavailability was tested using the audiogenic mouse test (Audiogenic seizures, R.L. Collins; Chapter 14, pages: 347-372. In: Experimental Models of Epilepsy; By: Pupura, Penny, Tower, Woodbury, Walter, Raven Press, New York, 1972. Standard Book Number: 0- 91 2 6-26-X) and/or the MES test. Where the MES test was used (as described below), the result is given in Table 8.
  • a compound could give 0%, 20%, 40%, 60%, 80% or 100% protection.
  • Compounds of the invention were given at a dose of 50 mg/kg, p.o., 1 hour prior to induction of convulsions (i.e. "pre-treatment time -1h").
  • ED50 values (ED: effective dose) were calculated using GraphPad Prism, v4.02.
  • mice blood was collected for determination of compounds' blood exposure. The results are shown below in Table 8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Pain & Pain Management (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
EP11757513.4A 2011-09-07 2011-09-07 Verwendung von 1h-chinazolin- 2,4 -dionen zur verwendung bei der prävention oder behandlung von lichtempfindlicher epilepsie Withdrawn EP2753331A1 (de)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2011/050687 WO2013036224A1 (en) 2011-09-07 2011-09-07 Use of 1h-quinazoline- 2, 4 -diones for use in the prevention or treatment photosensitive epilepsy

Publications (1)

Publication Number Publication Date
EP2753331A1 true EP2753331A1 (de) 2014-07-16

Family

ID=44652020

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11757513.4A Withdrawn EP2753331A1 (de) 2011-09-07 2011-09-07 Verwendung von 1h-chinazolin- 2,4 -dionen zur verwendung bei der prävention oder behandlung von lichtempfindlicher epilepsie

Country Status (10)

Country Link
EP (1) EP2753331A1 (de)
JP (1) JP2014525474A (de)
KR (1) KR20140071405A (de)
CN (1) CN103889427A (de)
AU (1) AU2011376333A1 (de)
BR (1) BR112014005210A2 (de)
CA (1) CA2846503A1 (de)
IN (1) IN2014DN01791A (de)
MX (1) MX2014002693A (de)
WO (1) WO2013036224A1 (de)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201609719A (zh) * 2014-05-28 2016-03-16 美國禮來大藥廠 作為TARP-γ8依賴性AMPA受體拮抗劑之6-經取代-3H-1,3-苯并噻唑-2-酮化合物
JP6902033B2 (ja) 2015-12-30 2021-07-14 アダマス ファーマシューティカルズ, インコーポレイテッド 発作−関連障害の処置のための方法および組成物

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0128996D0 (en) 2001-12-04 2002-01-23 Novartis Ag Organic compounds
SE0201943D0 (sv) 2002-06-20 2002-06-20 Astrazeneca Ab New use
WO2005044267A1 (en) 2003-10-31 2005-05-19 Astrazeneca Ab Alkynes ii
CN1871001A (zh) 2003-10-31 2006-11-29 阿斯利康(瑞典)有限公司 炔烃ⅲ
EP1677789A1 (de) 2003-10-31 2006-07-12 AstraZeneca AB Alkyne i
US20070149547A1 (en) 2004-02-12 2007-06-28 Celine Bonnefous Bipyridyl amides as modulators of metabotropic glutamate receptor-5
GB0507298D0 (en) 2005-04-11 2005-05-18 Novartis Ag Organic compounds
GB0508319D0 (en) 2005-04-25 2005-06-01 Novartis Ag Organic compounds
AR058554A1 (es) 2005-12-20 2008-02-13 Novartis Ag Compuestos heterociclicos nitrogenados de 6 miembros sustituidos, metodos para su preparacion, composiciones farmaceuticas que los contienen y su uso en el tratamiento de enfermedades mediadas por mglur5.
ES2753141T3 (es) * 2009-11-03 2020-04-07 Lupin Ltd Formulación de liberación modificada de lacosamida

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2013036224A1 *

Also Published As

Publication number Publication date
CN103889427A (zh) 2014-06-25
IN2014DN01791A (de) 2015-05-15
KR20140071405A (ko) 2014-06-11
MX2014002693A (es) 2014-06-04
JP2014525474A (ja) 2014-09-29
CA2846503A1 (en) 2013-03-14
BR112014005210A2 (pt) 2017-03-21
AU2011376333A1 (en) 2014-03-13
WO2013036224A1 (en) 2013-03-14

Similar Documents

Publication Publication Date Title
Trenité et al. Photosensitive epilepsy: a model to study the effects of antiepileptic drugs. Evaluation of the piracetam analogue, levetiracetam
US10111889B2 (en) Uses of ganaxolone
Rogawski et al. Preclinical pharmacology of perampanel, a selective non‐competitive AMPA receptor antagonist
DE60109589T2 (de) Pharmazeutische zusammensetzung zur behandlung von akutem schmerz, chronischem schmerz und/oder neuropathischem schmerz und migräne
AU2008271026A1 (en) A drug demonstrating anxiolytic effect based on hydrogenated pyrido (4, 3-B) indoles, its pharmacological compound and application method
JP6137833B2 (ja) 脱髄性および他の神経系疾患を患っている患者における神経認知的および/または神経精神医学的障害を改善するための4−アミノピリジンの使用
US7226949B2 (en) Compounds of use in the treatment of epilepsy, seizure, and electroconvulsive disorders
AU2016289271A1 (en) Therapeutic compounds and compositions for treating social disorders and substance use disorders
KR20170048541A (ko) 스미스 마제니스 증후군을 치료하기 위한 타시멜테온
EP2340254B1 (de) Zusammensetzungen und verfahren zur behandlung von epilepsie
WO2013036224A1 (en) Use of 1h-quinazoline- 2, 4 -diones for use in the prevention or treatment photosensitive epilepsy
CN111801096B (zh) 癫痫治疗剂
US20140163050A1 (en) Use of 1H-quinazoline-2,4-diones
EA023728B1 (ru) Терапевтическое средство от тревожных расстройств
Herrling et al. D-CPPene (SDZ EAA-494)—a competitive NMDA antagonist: pharmacology and results in humans
US20090270373A1 (en) Treatment of down syndrom with benzodiazepine receptor antagonists
CN111050800B (zh) 血清素3受体激动剂对疼痛的治疗
KR20120052341A (ko) 신경 세로이드 리포푸스신증의 치료에 사용하기 위한 1h-퀴나졸린-2,4-디온
BR112021004938A2 (pt) métodos e composições para tratamento de danos associados com o envelhecimento usando inibidores de ccr3
Hemanthkumar Study of anticonvulsant activity of acetazolamide on albino rats and its influence on anticonvulsant activity of sodium valproate
Kamath Study of Anticonvulsant effect of Simvastatin in Maximal Electroshock and Pentylenetetrazole Induced Seizure Model In Albino Mice
JP2013529622A (ja) 1h−キナゾリン−2,4−ジオンの使用
ZA200509646B (en) Further therapeutic use of zolpidem

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20140407

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20150414

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20150825