WO2020095979A1 - Therapeutic agent for central nervous system disease including tipepidine - Google Patents

Therapeutic agent for central nervous system disease including tipepidine Download PDF

Info

Publication number
WO2020095979A1
WO2020095979A1 PCT/JP2019/043606 JP2019043606W WO2020095979A1 WO 2020095979 A1 WO2020095979 A1 WO 2020095979A1 JP 2019043606 W JP2019043606 W JP 2019043606W WO 2020095979 A1 WO2020095979 A1 WO 2020095979A1
Authority
WO
WIPO (PCT)
Prior art keywords
tipepidine
disorder
pharmaceutical composition
acceptable salt
central
Prior art date
Application number
PCT/JP2019/043606
Other languages
French (fr)
Japanese (ja)
Inventor
剛志 辻村
瞬 林
Original Assignee
大日本住友製薬株式会社
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 大日本住友製薬株式会社 filed Critical 大日本住友製薬株式会社
Publication of WO2020095979A1 publication Critical patent/WO2020095979A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4525Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4535Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom, e.g. pizotifen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/49Cinchonan derivatives, e.g. quinine
    • 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
    • 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
    • A61P25/14Drugs 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a therapeutic agent for central diseases containing tipepidine.
  • Tipepidine is widely used in Japan as a central antitussive. Tipepidine is a cough medicine approved in 1959 and is a commonly used safe drug for pediatric patients. Tipepidine has a wide range of pharmacological actions, including central antitussive action, mood disorder, pain, schizophrenia, dysuria associated with cerebral infarction, attention deficit / hyperactivity disorder, cerebral dysfunction caused by environmental chemicals, Alzheimer's disease, It is also reported to be effective for obsessive-compulsive disorder. Further, it has been clarified from a small-scale clinical study that it is effective against attention deficit / hyperactivity disorder and depression (Patent Documents 1, 2, 3, 4, 5, Non-Patent Documents 1 and 2).
  • tipepidine has a very short half-life in vivo of 1.8 hours, and it is necessary to take it three times a day. Therefore, when used in a central disease, there is a problem that it is difficult to maintain the blood concentration, and furthermore, the intracerebral concentration that exhibits a drug effect cannot be sufficiently reached (Non-patent Document 3).
  • Non-Patent Documents 4 and 5 Non-Patent Documents 4 and 5).
  • Non-Patent Document 6 Non-Patent Document 6
  • Depression is classified as one of mood disorders, and one of the most common mental illnesses with a lifetime prevalence rate of 10% or higher.
  • the main symptoms are strong depressive mood, decreased motivation / interest / joy, psychomotor disorders (frustration / inhibition of mental activity), decreased appetite, insomnia, etc., and significantly deteriorates the quality of life of patients. ..
  • the treatment of depression includes cognitive-behavioral therapy, drug therapy, and electroconvulsive therapy (ECT), and drug therapy with antidepressants is actively used for moderate to moderate depression.
  • Antidepressants include tricyclic antidepressants (TCA), tetracyclic antidepressants, selective serotonin reuptake inhibitors (SSRI), serotonin / noradrenaline reuptake inhibitors, noradrenergic / selective serotonergic
  • TCA tricyclic antidepressants
  • SSRI selective serotonin reuptake inhibitors
  • NaSSA antidepressants
  • these drugs take 3 weeks or more for the onset of antidepressant action, and there is a problem that only about one-third of patients reach remission with the initial monotherapy.
  • ADHD Attention deficit / hyperactivity disorder
  • ICD10 ICD10
  • ICD10 ICD10
  • the main symptoms are hyperactivity, impulsivity, and carelessness, which make life difficult in school.
  • adolescence it causes severe mental illness such as depression and addiction, and significantly deteriorates the quality of life of the patient.
  • central stimulants dopain / noradrenaline reuptake inhibitors
  • non-central stimulants noradrenaline reuptake inhibitors, ⁇ 2 adrenergic agonists
  • central stimulants dopain / noradrenaline reuptake inhibitors
  • non-central stimulants noradrenaline reuptake inhibitors, ⁇ 2 adrenergic agonists
  • the central stimulants have a strong effect of suppressing the symptoms of ADHD, but have a risk of forming dependence and may be abused.
  • a non-central stimulant has a low risk of forming dependence, but a problem is that the effect of suppressing the symptoms of ADHD is weak. Therefore, there is a demand for a drug that can suppress ADHD symptoms to the same level as existing central stimulants without forming dependence.
  • it is also required to alleviate the symptoms of ADHD by using it together with a non-central stimulant or a central stimulant.
  • JP-A-2009-227631 Japanese Patent Laid-Open No. 2011-246446 JP2012-62272A Japanese Patent Laid-Open No. 2013-63958 JP, 2017-36242, A
  • Neuropsychiatric Disease and Treatment 2014, 10, 10, 147-151 Neuropsychiatric Disease and Treatment, 2014, 10, 10, 719-722 Asverin Tablets Drug Interview Form Experimental Physiology, 2007, 92, 67-78 Movement Disorders, 2001, 16, 16, 507-510 Neurology, 2000, 54, 1166-11756
  • the problem to be solved by the present invention is to provide a method and a medicine for treating or preventing central diseases caused by tipepidine.
  • the present invention relates to a therapeutic or prophylactic agent for central diseases, a method for treating or preventing central diseases, characterized in that tipepidine or a pharmaceutically acceptable salt thereof is used in combination with a CYP2D6 inhibitor.
  • Pharmaceutical compositions and kits are included in the present invention.
  • the present invention also includes tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotic drugs, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating.
  • the present invention provides a therapeutic or prophylactic agent for addiction, fibromyalgia, or a group of trauma and stress-related disorders, a method for treating or preventing, a pharmaceutical composition and a kit.
  • the present invention relates to the following.
  • Item 1 A pharmaceutical composition for treating or preventing a central disease, which comprises tipepidine or a pharmaceutically acceptable salt thereof in combination with a CYP2D6 inhibitor.
  • Item 2 A pharmaceutical composition for treating or preventing a central disease, comprising a CYP2D6 inhibitor, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
  • Item 3 A pharmaceutical composition for treating or preventing a central disease, comprising tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
  • CYP2D6 inhibitors are quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, duloxetine, mirabegron, celecoxib, escitalopram, moclobelatil, clavitone, fluvoxamine, abiraterabalone, mirabedalone, amiodarone, fluvoxamine, abiraterabalone, amiodalone, amiodala. 4.
  • composition according to any one of the above items 1 to 3, which is one or more drugs selected from the group consisting of, ritonavir, sertraline, vemurafenib, and deramciclane, or a pharmaceutically acceptable salt thereof. ..
  • the CYP2D6 inhibitor is one or more pharmaceutical agents selected from the group consisting of quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, duloxetine, mirabegron, celecoxib, escitalopram, moclobemide, cimetidine, and fluvoxamine; Item 4.
  • the pharmaceutical composition according to any one of Items 1 to 3, wherein the pharmaceutical composition is a salt acceptable for.
  • CYP2D6 inhibitor is one or more drugs selected from the group consisting of quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, and dacomitinib, or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition according to any one of claims.
  • the CYP2D6 inhibitor is one or more drugs selected from the group consisting of quinidine, paroxetine, bupropion, and fluoxetine, or a pharmaceutically acceptable salt thereof, according to any one of items 1 to 3 above.
  • Pharmaceutical composition is one or more drugs selected from the group consisting of quinidine, paroxetine, bupropion, and fluoxetine, or a pharmaceutically acceptable salt thereof, according to any one of items 1 to 3 above.
  • Item 8 The pharmaceutical composition according to any one of Items 1 to 3, wherein the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof.
  • Central diseases are Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central diseases, overeating, addiction, or fibromyalgia, the above-mentioned item
  • Item 10 The pharmaceutical composition according to any one of Items 1 to 8, wherein the central disease is a sleep disorder.
  • Item 11 The pharmaceutical composition according to any one of items 1 to 8 and 10, wherein the central disease is hypersomnia, idiopathic hypersomnia disorder, or narcolepsy.
  • Item 12. The pharmaceutical composition according to any one of items 1 to 8, wherein the central disease is a depressive disorder group.
  • Central disorders include anxiety-related depressive disorders, depressive disorders with mixed features, depressive disorders with melancholic features, depressive disorders with mood-matching psychotic features, mood Depressive disorders with inconsistent psychotic features, depressive disorders with catatonic disorders, seasonal depressive disorders, severe dysregulation, depression, persistent depressive disorder, premenstrual dysphoric disorder, or substances / medicines 13.
  • the pharmaceutical composition according to any one of the above items 1 to 8 and 12, which is an induced depressive disorder.
  • Item 14 The pharmaceutical composition according to any one of items 1 to 8 and 12, wherein the central disease is a group of depressive disorders in children.
  • Central disorders are anxiety-affected depressive disorders in children, depressive disorders with mixed features in children, depressive disorders with melancholic features in children, psychotic features that match mood in children Depressive disorder group with illness, depressive disorder group with psychotic features inconsistent with mood in children, depressive disorder group with catatonic disorders in children, seasonal depressive disorder group in children, severe mood regulation in children, in children 15.
  • Item 16 9. The pharmaceutical composition according to any one of items 1 to 8 above, wherein the central disease is an anxiety group.
  • Item 17. 17 The pharmaceutical composition according to any one of the above items 1 to 8 and 16, wherein the central disease is separation anxiety disorder, localized phobia, social anxiety disorder, panic disorder, or substance / drug-induced anxiety disorder. ..
  • Item 18. The pharmaceutical composition according to any one of items 1 to 8 and 16 above, wherein the central disease is an anxiety group in children.
  • the central disease is separation anxiety in children, localized phobia in children, social anxiety in children, panic disorder in children, or substance / drug-induced anxiety in children.
  • the pharmaceutical composition according to any one of claims.
  • Item 20 The pharmaceutical composition according to any one of Items 1 to 8, wherein the central disease is a neurodevelopmental group.
  • Item 21 Central diseases include autism spectrum disorder, attention deficit / hyperactivity disorder, localized learning disorder, intellectual disability, speech disorders, speech disorders, childhood-onset fluency, social (pragmatic) communication disorders, development 21.
  • Item 22 The pharmaceutical composition according to any one of items 1 to 8 above, wherein the central disease is a group of obsessive-compulsive disorder and related disorders.
  • Item 23 The pharmaceutical composition according to any one of the above items 1 to 8 and 22, wherein the central disease is obsessive-compulsive disorder, dysphoric phobia, dentosis, hair loss, or plucking skin.
  • Item 24 The pharmaceutical composition according to any one of items 1 to 8, wherein the central disease is a group of disorders related to trauma and stress factors.
  • Item 25 The medicine according to any one of the above items 1 to 8 and 24, wherein the central disease is post-traumatic stress disorder, reactive attachment disorder, disinhibition-type interpersonal interaction disorder, acute stress disorder, or adaptation disorder. Composition.
  • Item 26 The pharmaceutical composition according to any one of the above items 1 to 25, wherein the dose of tipepidine or a pharmaceutically acceptable salt thereof is 2 mg to 2000 mg per day.
  • Item 27 The pharmaceutical composition according to any one of items 1 to 26 above, wherein the dose of the CYP2D6 inhibitor is 1 mg to 1000 mg per day.
  • Item 28 A method for treating or preventing a central disease comprising administering to a mammal a therapeutically effective amount of tipepidine or a pharmaceutically acceptable salt thereof and a therapeutically effective amount of a CYP2D6 inhibitor.
  • Item 29 Tipepidine or a pharmaceutically acceptable salt thereof for use in the treatment or prevention of central diseases in combination with a CYP2D6 inhibitor.
  • Item 30 A CYP2D6 inhibitor for use in the treatment or prevention of central diseases, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
  • Item 31 Use of tipepidine or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing central diseases, which is used in combination with a CYP2D6 inhibitor.
  • Item 32 Use of a CYP2D6 inhibitor for the manufacture of a medicament for treating or preventing central diseases, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
  • Item 33 Use of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor for the manufacture of a medicament for treating or preventing central diseases.
  • Item 34 A kit for the treatment or prevention of central diseases, which comprises tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
  • Parkinson's disease due to antipsychotic use, sleep disorder, chronic fatigue syndrome, fatigue associated with neurodegeneration and central illness, overeating, addiction, fiber containing tipepidine or a pharmaceutically acceptable salt thereof
  • Item 36 Including tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorder, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating, addiction, or A therapeutic or preventive agent for fibromyalgia.
  • Item 37 The therapeutic or prophylactic agent according to Item 35 or 36, wherein the disease is a sleep disorder.
  • Item 39 The therapeutic or prophylactic agent according to Item 35 or 36 above, wherein the disease is a group of trauma and stress-related disorders.
  • Item 40 37.
  • the therapeutic or prophylactic agent according to the above item 35 or 36, wherein the disease is post-traumatic stress disorder, reactive attachment disorder, disinhibition-type interpersonal interaction disorder, acute stress disorder, or adaptation disorder.
  • the invention also relates to: Fatigue associated with Parkinson's disease, Parkinson's disease syndrome due to antipsychotic drug use, sleep disorders, chronic fatigue syndrome, neurodegeneration and central illness, including administration of tipepidine or a pharmaceutically acceptable salt thereof to a mammal , A method for treating or preventing hyperphagia, addiction, fibromyalgia, or trauma and stress-related disorders.
  • Parkinson's disease Parkinson's syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central illness, overeating, addiction, fibromyalgia, or trauma and stress-related disorders
  • a pharmaceutically acceptable salt thereof for use in the treatment or prophylaxis of Parkinson's disease, Parkinson's syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central illness, overeating, addiction, fibromyalgia, or trauma and stress-related disorders
  • tipepidine or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prophylaxis; and if the disease is a sleep disorder (especially hypersomnia, idiopathic hypersomnia disorder, or narcolepsy).
  • the present invention by combining tipepidine or a pharmaceutically acceptable salt thereof with a CYP2D6 inhibitor, the pharmacokinetics of tipepidine having a short half-life in vivo were improved, and a new central action of tipepidine was found.
  • Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotics, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating, dependence, or therapeutic agents for central diseases such as fibromyalgia
  • the present invention can be expected to improve sleep disorders.
  • tipepidine or a pharmaceutically acceptable salt thereof comprising tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotic drugs, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating.
  • therapeutic or prophylactic agents for addiction, or fibromyalgia methods for treating or preventing, pharmaceutical compositions and kits.
  • tipepidine or a pharmaceutically acceptable salt thereof in combination with a CYP2D6 inhibitor, a depressive disorder group, anxiety group, neurodevelopmental group, obsessive-compulsive and related disease group, or heart.
  • therapeutic agents or prophylactic agents for the group of trauma and stress-related disorders methods for treating or preventing, pharmaceutical compositions and kits.
  • a therapeutic or prophylactic agent for a group of disorders related to trauma and stress factors a method, a pharmaceutical composition and a kit for treating or preventing, comprising tipepidine or a pharmaceutically acceptable salt thereof.
  • FIG. 1 shows the metabolic stability of tipepidine in human hepatocytes, and shows the residual rates of tipepidine in the treated and non-treated groups to which 1-ABT, a CYP inhibitor, was added.
  • the horizontal axis shows the reaction time, and the vertical axis shows the residual ratio of tipepidine.
  • FIG. 2 shows simulated changes in plasma concentration of tipepidine when orally administered to humans alone with tipepidine or with combined use of a CYP2D6 inhibitor and tipepidine.
  • the horizontal axis shows the time from administration, and the vertical axis shows the plasma concentration of tipepidine.
  • FIG. 1 shows the metabolic stability of tipepidine in human hepatocytes, and shows the residual rates of tipepidine in the treated and non-treated groups to which 1-ABT, a CYP inhibitor, was added.
  • the horizontal axis shows the reaction time, and the vertical axis shows the residual ratio of tipepidine.
  • FIG. 2 shows simulated changes in
  • FIG. 3 shows plasma of tipepidine when orally administered to human hepatocyte-transplanted mice (PXB mice) with 20 mg / kg of tipepidine hibenzate alone and with 100 mg / kg of CYP2D6 inhibitor quinidine and 20 mg / kg of tipepidine hibenzate.
  • the figure shows the transition of medium concentration.
  • the horizontal axis shows the time from administration, and the vertical axis shows the plasma concentration of tipepidine.
  • FIG. 4 shows changes in plasma concentrations of tipepidine when SD rats were orally administered with 20 mg / kg of tipepidine hibenzate alone, and combined with 20 mg / kg of CYP inhibitor 1-ABT and 100 mg / kg of tipepidine hibenzate. It is a thing.
  • FIG. 5 shows the total amount of locomotor activity from 30 minutes after administration to 1.5 hours after intraperitoneal administration of solvent or tipepidine hibenzate (2 groups of 25 mg / kg and 80 mg / kg) to SD rats. It was done.
  • the bar graph shows the average value of each treatment group, and ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01.
  • FIG. 6 shows that SD rats were intraperitoneally administered with a solvent, imipramine 10 mg / kg was subcutaneously administered, or tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg) was intraperitoneally administered 30 minutes later.
  • the forced swimming test was performed, and the immobility time (seconds) of the rat was displayed.
  • the bar graph shows the average value of each treatment group, and ** means that the risk rate (P) calculated by the Tukey-Kramer test is less than 0.01.
  • FIG. 7 shows that SD rats were forcibly orally administered with vehicle, tipepidine hibenzate 20 mg / kg, or tipepidine hibenzate 20 mg / kg and CYP inhibitor 1-ABT 100 mg / kg, and 30 minutes after the administration of tipepidine, 1.5 hours. It is a display of the total sum of the spontaneous locomotor activity of.
  • the bar graph shows the average value of each treatment group.
  • S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more.
  • FIG. 8 shows that SD rats were forcibly orally administered with solvent, tipepidine hibenzate 20 mg / kg, or tipepidine hibenzate 20 mg / kg and CYP inhibitor 1-ABT 100 mg / kg, and a forced swimming test was conducted 1 hour after the administration. , The immobility time of the rat (second) is displayed. The bar graph shows the average value of each treatment group.
  • S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more, and ** means that the risk factor (P) calculated by the Tukey-Kramer test is less than 0.01.
  • FIG. 10A shows that Wistar rats were intraperitoneally administered with a solvent, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg), sleep EEG was measured 4 hours after the administration, and the awakening time of the rats was measured. It is displayed.
  • FIG. 10B is a graph showing the total wakeup time 4 hours after the administration of the solvent, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg) to the Wistar rat, intraperitoneally.
  • the bar graph shows the average value of each treatment group, ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01, and * indicates the risk rate (P) calculated by Dunnett's test. Means less than 0.05.
  • FIG. 11A shows that Wistar rats were intraperitoneally administered with a solvent, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg). REM sleep EEG was measured 4 hours after administration, and the REM sleep time of the rat was displayed.
  • FIG. 11B is a graph showing the total REM sleep time of 4 hours after the intraperitoneal administration of the vehicle, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg), to Wistar rats.
  • the bar graph shows the average value of each treatment group, ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01, and * indicates the risk rate (P) calculated by Dunnett's test. Means less than 0.05.
  • FIG. 12 shows the immobility time by the forced swimming test 60 minutes after the administration, which was subcutaneously administered to a chronically fatigued SD rat at a dose of 1 mg / kg of solvent and tipepidine citrate or 5 mg / kg. It is a thing. ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01.
  • FIG. 13A shows that on the day before the test, Wistar rats were forcibly orally administered with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the solvent or tipepidine hibenzate 40 mg / kg was orally administered, and the test was started. The sleep electroencephalogram was measured 6 hours later, and the awakening time of the rat was displayed.
  • FIG. 13A shows that on the day before the test, Wistar rats were forcibly orally administered with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the solvent or tipepidine hibenzate
  • FIG. 13B shows that on the day before the test, Wistar rats were gavaged with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the vehicle or the tipepidine hibenzate 40 mg / kg was gavaged, and the test was started. It shows the total awakening time of the next 6 hours.
  • the bar graph shows the average value of each treatment group.
  • S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more, and ** means that the risk factor (P) calculated by the Tukey test is less than 0.01.
  • FIG. 14A shows that on the day before the test, Wistar rats were gavaged with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the vehicle or the tipepidine hibenzate salt 40 mg / kg was gavaged orally, and the test was started. The REM sleep electroencephalogram was measured 6 hours later, and the REM sleep time of the rat was displayed.
  • FIG. 14B shows that on the day before the test, Wistar rats were gavaged with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the vehicle or the tipepidine hibenzate 40 mg / kg was gavaged, and the test was started.
  • One aspect of the present invention relates to a combination of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
  • Another aspect of the present invention comprises tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central diseases.
  • the present invention relates to a therapeutic or preventive agent for feeling, overeating, addiction, or fibromyalgia.
  • Tipepidine in the present invention has a chemical name of 3- (dithien-2-ylmethylene) -1-methylpiperidine and has already been used as an active ingredient of a commercially available antitussive drug. And, as tipepidine, it can be used in a free base form or a pharmaceutically acceptable acid addition salt and / or hydrate and / or solvate thereof (in the present specification, these are collectively referred to as tipepidine Or its pharmaceutically acceptable salt).
  • Suitable acid addition salts are, for example, selected from succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulfonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulfuric acid, tartaric acid, citric acid and hibenzic acid. Examples thereof include salts of acids, preferably salts of hibenzic acid or citric acid.
  • cytochrome P450 (hereinafter referred to as CYP) is known as an enzyme that plays a major role.
  • CYP cytochrome P450
  • the major molecular species are CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A4 / 5. Since the CYP2D6 inhibitor has an action of suppressing the enzymatic activity of CYP2D6, it increases blood exposure of the CYP2D6 substrate drug when used in combination with a drug metabolized by CYP2D6 (hereinafter referred to as CYP2D6 substrate drug).
  • the “CYP2D6 inhibitor” in the present invention means a plasma exposure (AUC) of a CYP2D6 substrate drug susceptible to interaction in a clinical drug interaction test by 1.25 times or more, preferably 2 times or more, more preferably 5 times or more. It means a drug having an inhibitory ability to increase more than twice.
  • CYP2D6 inhibitor in the present invention, specifically, “Drug Interaction Guideline for Drug Development and Proper Information Provision (Final Draft)” issued on July 8, 2014 or September 26, 2016 Drug Development and Drug Interactions: Table of of Substrates, Inhibitors and and Inducers Table 3-2: Examples of clinical inhibitors for P450-mediated metabolisms (for concomitant use priorical and D quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, which are classified as strong inhibitors of CYP2D6 in drug labeling), duloxetine, mirabegron, celecoxib, escitalopram, moclobemide, which are classified as moderate inhibitors.
  • CYP2D6 inhibitor any two or more of these drugs may be used in combination. Although these drugs have different main actions, they also have a CYP2D6 inhibitory action, and are therefore referred to as “CYP2D6 inhibitors” in the present specification.
  • Suitable acid addition salts are selected, for example, from succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulfonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulfuric acid, tartaric acid, citric acid and hibenzic acid. Acid salts may be mentioned.
  • quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib or pharmaceutically acceptable salts thereof which are classified as strong inhibitors of CYP2D6, and more preferably have few side effects and are easy to clinically Quinidine, paroxetine, bupropion, fluoxetine, or a pharmaceutically acceptable salt thereof, wherein quinidine is a sulfate hydrate, paroxetine is a hydrochloride hydrate, bupropion is a hydrochloride, and fluoxetine is a hydrate. It is preferably used as the hydrochloride salt. Even more preferably, quinidine or a pharmaceutically acceptable salt thereof, which is different from the effective dose exhibiting the main drug effect by 10 times or more the effective dose exhibiting the inhibitory effect, is exemplified.
  • Parkinson's disease As one embodiment of the central diseases in the present invention, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotic agents, sleep disorders (including hypersomnia, idiopathic hypersomnia disorder, narcolepsy), chronic fatigue syndrome, neurodegeneration and central nervous system Fatigue associated with sexual disorders, overeating, addiction, or fibromyalgia.
  • sleep disorders including hypersomnia, idiopathic hypersomnia disorder, narcolepsy
  • chronic fatigue syndrome neurodegeneration and central nervous system Fatigue associated with sexual disorders, overeating, addiction, or fibromyalgia.
  • Preferred examples include hypersomnia, idiopathic hypersomnia disorder, or sleep disorders including narcolepsy.
  • DSM-5 Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition
  • DSM-5 Depressive disorders
  • category V anxiety Anxiety Disorders
  • category I neurodevelopmental disorders Neurodevelopmental disorders
  • Depressive Disorders include Depressive Disorders With With anxious distress, Depressive Disorders With Mixed features, and depression with melancholic features. Depressive Disorders With melancholicfeatures, Depressive Disorders With-moog-congruent psychoticfeatures, Depressive Disorders with non-mood psychotic features With-mood-incongruent psychotic features, Depressive Disorders With Catatonia, Depressive Disorders, With seasonal pattern, Severe Mood Dysregulation Disorder, Depression.
  • Depressive Disorder Major Depressive Disorder
  • persistent depressive disorder dysthymia
  • disiste nt Depressive Disorder Dysthymia
  • premenstrual dysphoric disorder Premenstrual Dysphoric Disorder
  • substance / drug-induced depressive disorder Substance / Medication-Induced Depressive Disorder
  • Anxiety Disorders include Separation Anxiety Disorder, Specific Phobia, Social Anxiety Disorder (Social Phobia), and Panic Disorder. ), Or substance / drug-induced anxiety disorder (Substance / Medication-Induced Anxiety Disorder).
  • Neurodevelopmental Disorders include Autism Spectrum Disorder, Attention-Deficit / Hyperactivity Disorder, Specific Learning Disorder, and intellectual disability IntellectualDisability), Language Disorder, Speech Sound Disorder, Childhood-Onset Fluency Disorder (Stuttering), Social (Pragmatic) Communication (Pragmatic) Communication Disorder), Developmental Coordination Disorder, Tic Disorders.
  • depression disorders in children include depressive disorders with anxious distress in children, depressive disorders with mixed features in children, depressive disorders with melancholic features in children, and psychosis consistent with mood in children.
  • Depressive disorder group with sexual characteristics depressive disorder group with psychotic features that do not match mood in children, depressive disorder group with catatonic disorders in children, seasonal depressive disorder group in children, severe mood regulation in children , Depression in children, persistent depressive disorder in children (dysthymia), premenstrual dysphoric disorder in children, or substance-drug-induced depressive disorder in children.
  • Anxiety groups in children include isolated anxiety in children, localized phobia in children, social anxiety in children (social phobia), panic disorder in children, or substance-drug-induced anxiety in children.
  • the term “children” used in the present invention generally means 6 to 15 years old.
  • the therapeutic or prophylactic agent for a depressive disorder group, anxiety group, or neurodevelopmental group by combining tipepidine or a pharmaceutically acceptable salt thereof with a CYP2D6 inhibitor according to the present invention has immediate effect. It is a safe medicine that is effective, persistent, does not form addiction, and does not cause suicide attempts, and is therefore applicable not only to adult patients but also to pediatric patients.
  • a group VI according to DSM-5 is obsessive-compulsive and related disorders (Obsessive-Compulsive and Related Related Disorders), or a group VII of trauma and stress-related disorders. (Trauma- and Stressor-Related Disorders).
  • Obsessive-compulsive Disorder Obsessive-Compulsive Disorder, Body Dysmorphic Disorder, Hoarding Disorder, Trichotillomania (Hair-Pulling Disorder), or skin Exclusion (Skin-Picking) Disorder is mentioned.
  • Post-traumatic stress disorder including post-traumatic stress disorder in children under 6 years old
  • Posttraumatic StressDisorder reactive attachment disorder
  • Reactive AttachmentDisorder Disinhibited social interaction disorder
  • acute stress disorder acute Stress Disorder
  • adjustment disorder Adjustment Disorders
  • a "therapeutically effective amount” means an amount of a drug or drug that elicits a biological or pharmaceutical response required by a researcher or physician in a tissue, system, animal or human.
  • “treatment” and “treatment” refer to any treatment of a disease (eg, amelioration of symptoms, alleviation of symptoms, suppression of progression of symptoms, etc.) and any prevention of diseases (eg, onset and / or development of a disease). Or prevention of progress) is included.
  • pharmaceutical composition As used herein, “pharmaceutical composition,” “pharmaceutical formulation,” and “pharmaceutical product” refer to products that contain the specified ingredients in the specified amounts, as well as directly or indirectly from combinations of the specified ingredients in the specified amounts. It is intended to encompass any product that results.
  • the definition of sleep disorder used in the present invention is "a state in which excessive sleepiness or actual falling into sleep during the day is seen repeatedly every day, which lasts for at least one month, As a result, social life or occupational function is disturbed, or one feels distressed, even if the duration of one session is shorter than one month and repeated sleep periods are observed. ”
  • both agents may be administered separately or may be administered together as one pharmaceutical composition.
  • one component of the combination of the invention may be administered prior to, simultaneously with, or after the other component.
  • These components may be prepared as a pharmaceutical preparation in a single preparation form or separate preparation forms, or may be a kit.
  • the active ingredient of the present invention is orally or parenterally (for example, intramuscular, intraperitoneal, intravenous, transdermal or subcutaneous injection or implant).
  • parenterally for example, intramuscular, intraperitoneal, intravenous, transdermal or subcutaneous injection or implant.
  • it may be formulated alone or in combination with other drugs that can be used in combination in a suitable formulation containing a vehicle.
  • Pharmaceutical products suitable for oral administration according to the invention comprise the active compound tipepidine of the invention, a pharmaceutically acceptable salt thereof and / or a CYP2D6 inhibitor, in a dose adapted to the individual needs of the active ingredient. It can be administered orally in the usual dosage forms, such as tablets, capsules, syrups, suspensions and the like. Alternatively, the solution, emulsion, suspension or the like in the form of a liquid preparation can be administered parenterally in the form of injection, or can be made into a patch. Also, the above suitable dosage forms can be prepared by incorporating the active compound into an acceptable conventional carrier, excipient, binder, stabilizer and the like. When used in an injection dosage form, an acceptable buffering agent, solubilizing agent, isotonic agent, pH adjusting agent and the like can be added.
  • the dose of the active ingredient of the present invention is not particularly limited, but the dose and frequency of administration of the therapeutic agent vary depending on the administration form or the degree of disease state requiring treatment.
  • tipepidine or a pharmaceutically acceptable salt thereof is administered as a single agent, 2-2000 mg, preferably 20-200 mg per day for an adult is orally or parenterally administered once or in several divided doses. be able to.
  • tipepidine or a pharmaceutically acceptable salt thereof when tipepidine or a pharmaceutically acceptable salt thereof is administered in combination with a CYP2D6 inhibitor, the dose is 2-2000 mg, preferably 20-200 mg per day for an adult, orally or
  • the CYP2D6 inhibitor may be orally administered, and the combined CYP2D6 inhibitor may be 1-1000 mg, preferably 1-300 mg per day for an adult, and may be orally or parenterally administered once or in several divided doses.
  • quinidine or a pharmaceutically acceptable salt thereof can be administered orally or parenterally per day for an adult at 1 to 200 mg, preferably 1 to 50 mg, once or in several divided doses. ..
  • paroxetine or a pharmaceutically acceptable salt thereof can be orally or parenterally administered to an adult per day at 1 to 200 mg, preferably 1 to 50 mg, once or in several divided doses. ..
  • bupropion or a pharmaceutically acceptable salt thereof can be administered orally or parenterally in an amount of 1 to 1000 mg, preferably 1 to 300 mg per day for an adult, or once or in several divided doses. ..
  • fluoxetine or a pharmaceutically acceptable salt thereof can be administered orally or parenterally in an adult daily dose of 1-200 mg, preferably 1-50 mg, once or in several divided doses. ..
  • the weight ratio of tipepidine (free form equivalent): CYP2D6 inhibitor (free form equivalent) when administering in combination with tipepidine or a pharmaceutically acceptable salt thereof and the CYP2D6 inhibitor is 1: 500 to 2000: 1.
  • free form equivalent means the weight as it is when the main drug is used in the free form, and the free weight excluding the weight of the salt when the main drug is used in the form of a pharmaceutically acceptable salt.
  • the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof
  • tipepidine or a pharmaceutically acceptable salt thereof and quinidine or a pharmaceutically acceptable salt thereof are administered in combination.
  • the weight ratio of tipepidine (free body conversion): quinidine (free body conversion) is 1: 100 to 2000: 1.
  • the weight ratio of tipepidine (calculated as free form): paroxetine is 1: 100 to 2000: 1.
  • the CYP2D6 inhibitor is bupropion or a pharmaceutically acceptable salt thereof
  • tipepidine or a pharmaceutically acceptable salt thereof and bupropion or a pharmaceutically acceptable salt thereof are administered in combination.
  • the weight ratio of tipepidine (free form conversion): bupropion is 1: 500 to 2000: 1.
  • the CYP2D6 inhibitor is fluoxetine or a pharmaceutically acceptable salt thereof
  • tipepidine or a pharmaceutically acceptable salt thereof and fluoxetine or a pharmaceutically acceptable salt thereof are administered in combination.
  • the weight ratio of tipepidine (free form conversion): fluoxetine is 1: 100 to 2000: 1.
  • the lower limit value of tipepidine (free body conversion) per day for adults when tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor are administered in combination is in the range of 2 to 20 mg.
  • the lower limit of the daily dose of the CYP2D6 inhibitor for an adult is any weight in the range of 1 to 20 mg, for example, 1 mg, 2 mg, 5 mg, 10 mg. , Or 20 mg.
  • the lower limit of the weight ratio of tipepidine (free form conversion): CYP2D6 inhibitor is, for example, 1: 500, 1: 250, 1: 100, 1:50, 1:25, 1:10, 1: 5, 1: 2.
  • the CYP2D6 inhibitor when the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof, 1 mg, 2 mg, or 5 mg can be mentioned as the lower limit of quinidine (free form conversion) per day for an adult.
  • the lower limit of the weight ratio of tipepidine (converted free form): quinidine (converted free form) may be 1: 100, 1:50, 1:25, 1:10, 1: 5 or 1: 2.
  • the lower limit of paroxetine (free body conversion) for an adult per day is 1 mg, 2 mg, or 5 mg.
  • the lower limit of the weight ratio of tipepidine (converted to free form): paroxetine (converted to free form) includes 1: 100, 1:50, 1:25, 1:10, 1: 5 or 1: 2.
  • the CYP2D6 inhibitor is bupropion or a pharmaceutically acceptable salt thereof
  • 1 mg, 10 mg, or 20 mg may be mentioned as the lower limit of bupropion (free form conversion) per day for an adult.
  • the lower limit of the weight ratio of tipepidine (converted free form): bupropion (converted free form) includes 1: 500, 1: 300, 1: 150, 1: 100, 1:50 or 1:25.
  • the lower limit of fluoxetine (free form conversion) per day for an adult is 1 mg, 2 mg, or 5 mg.
  • the lower limit of the weight ratio of tipepidine (free form conversion): fluoxetine (free form conversion) may be 1: 100, 1:50, 1:25, 1:10, 1: 5 or 1: 2.
  • the upper limit of the amount of tipepidine (free body conversion) per day for an adult when the tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor are administered in combination is in the range of 200 to 2000 mg.
  • the upper limit of the daily dose of the CYP2D6 inhibitor for an adult is any weight in the range of 50 to 1000 mg, for example, 50 mg, 100 mg, 200 mg, 500 mg. , Or 1000 mg.
  • the upper limit of the weight ratio of tipepidine (free form conversion): CYP2D6 inhibitor is, for example, 2000: 1, 1000: 1, 800: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1.
  • the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof, 50 mg, 100 mg, or 200 mg may be mentioned as the upper limit of quinidine (free body conversion) per day for an adult.
  • the upper limit of the weight ratio of tipepidine (converted free form): quinidine (converted free form) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
  • the CYP2D6 inhibitor when the CYP2D6 inhibitor is paroxetine or a pharmaceutically acceptable salt thereof, 50 mg, 100 mg, or 200 mg may be mentioned as the upper limit of paroxetine (free form conversion) per day for an adult.
  • the upper limit of the weight ratio of tipepidine (converted free form): paroxetine (converted free form) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
  • the CYP2D6 inhibitor when the CYP2D6 inhibitor is bupropion or a pharmaceutically acceptable salt thereof, the upper limit of bupropion (free body conversion) for an adult per day is 300 mg, 600 mg, or 1000 mg.
  • the upper limit of the weight ratio of tipepidine (free form conversion): bupropion (free form conversion) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
  • the CYP2D6 inhibitor is fluoxetine or a pharmaceutically acceptable salt thereof
  • 50 mg, 100 mg, or 200 mg may be mentioned as the upper limit of fluoxetine (free body conversion) per day for an adult.
  • the upper limit of the weight ratio of tipepidine (free form conversion): fluoxetine (free form conversion) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
  • the pharmaceutical composition of the present invention is used as a solid preparation for oral administration, a liquid preparation for oral administration, and an injection, an external preparation, a suppository, an inhalant, a nasal preparation, etc. for parenteral administration.
  • the solid preparations for oral administration include tablets, pills, capsules, powders and granules. Capsules include hard capsules and soft capsules. In addition, tablets include sublingual tablets, buccal tablets, and rapidly disintegrating buccal tablets.
  • one or more active substances may be used as they are, or excipients (lactose, mannitol, glucose, crystalline cellulose, dextrin, starch, etc.), binders (hydroxypropylcellulose, polyvinylpyrrolidone). , Magnesium metasilicate, etc.), disintegrant (calcium fibrin glycolate, etc.), lubricant (magnesium stearate, talc, etc.), stabilizer, solubilizer (glutamic acid, aspartic acid, etc.), etc. It is used by formulating it according to a conventional method.
  • a coating agent sucrose, gelatin, hydroxypropylcellulose, hydroxypropylmethylcellulose phthalate, etc.
  • capsules of absorbable material such as gelatin.
  • additives such as preservatives, antioxidants, colorants, sweeteners and the like which are commonly used can be added if necessary.
  • Sublingual tablets, buccal patches, rapidly disintegrating buccal tablets, and oral liquid preparations for oral administration are mixed with usual carriers, excipients, binders, stabilizers, etc. suitable for each dosage form. It is manufactured according to a known method.
  • the dosage form of the external preparation for parenteral administration includes, for example, ointments, gels, creams, poultices, patches, liniments, sprays, inhalants, sprays, aerosols, eye drops, and Includes nasal drops and the like. These contain one or more active substances and are produced by a known method or a commonly used formulation.
  • a poultice, a patch, and an injection for parenteral administration are manufactured by a known or commonly used formulation by mixing with a usual pharmaceutical additive suitable for each dosage form.
  • the term “effective amount” refers to tipepidine or a pharmaceutical preparation thereof, which completely or partially inhibits the progression of central diseases, or at least partially alleviates one or more symptoms of central diseases. Or a combination of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
  • An effective amount can be a therapeutically or prophylactically effective amount.
  • An effective amount will be determined by the age and sex of the patient, the condition being treated, the severity of the condition, the results sought, and the like. For a given patient, the effective amount can be determined by methods known to those of ordinary skill in the art.
  • Example 1 Estimation of CYP metabolism contribution of tipepidine using human hepatocytes The contribution of CYP metabolism to the metabolism of tipepidine by human hepatocytes was confirmed by the following method.
  • Human frozen hepatocytes used were those manufactured by Sekisui XenoTech. Human frozen hepatocytes were seeded in KHB buffer at a concentration of 1 million cells / mL, and tipepidine was added thereto at a concentration of 1 ⁇ mol / L.
  • a treatment group to which 1-aminobenzotriazole (1-ABT) (2.5 mmol / L), which is a CYP inhibitor, was added and a non-treatment group to which no CYP inhibitor was added were provided, and the conditions were 37 ° C. and 5% CO 2 And incubated for 1 hour or 2 hours.
  • the residual rate of tipepidine after the reaction was measured by LC-MS.
  • Example 2 Calculation of Various CYP Metabolism Contribution Ratios of Tipepidine Using Human Liver Microsomes
  • the metabolic contribution ratios of various CYPs involved in the metabolism of tipepidine were calculated using human liver microsomes.
  • Human liver microsomes used were manufactured by Sekisui XenoTech. Human liver microsomes were added at a concentration of 0.1 mg / mL, NADPH at 3 mmol / L, and tipepidine at a concentration of 0.1 ⁇ mol / L in a 50 mmol / L phosphate buffer (pH 7.4), and various CYPs were added.
  • the selective inhibitors were mixed at the concentrations shown below and incubated at 37 ° C for 30 minutes.
  • CYP1A2 Concentration of selective inhibitor of various CYPs Furafylline (CYP1A2): 10 ⁇ mol / L, montelukast (CYP2C8): 10 ⁇ mol / L, sulfaphenazole (CYP2C9): 10 ⁇ mol / L, N-benzylnilvanol ( N-benzylnirvanol; CYP2C19): 5 ⁇ mol / L, quinidine (CYP2D6): 1 ⁇ mol / L, azamulin (CYP3A4): 5 ⁇ mol / L
  • Table 1 shows the survival rate at the time of inhibition of various CYPs in the metabolism of tipepidine in human liver microsomes, the metabolic clearance, and the metabolic contribution rate of various CYPs calculated from the metabolic clearance. For the first time, it was revealed that tipepidine was mainly metabolized by CYP2D6, and its contribution rate was calculated to be 83%.
  • Example 3 Simulation of Tipepidine Plasma Concentration Changes in Humans and PK Parameters in Combination with Tipepidine Alone, CYP2D6 Inhibitor and Tipepidine Combined Commercially-Containing Tipepidine as an Active Ingredient in Plasma Concentration Changes and PK Parameters after Oral Administration of Tipepidine It was calculated by applying the information described in the Asverin Tablet Interview Form (IF) to the following 1-compartment model formula.
  • IF Asverin Tablet Interview Form
  • FIG. 2 shows the simulation results of changes in the plasma concentration of tipepidine when orally administered to humans alone with tipepidine alone, or when orally administered with the combination of CYP2D6 inhibitor and tipepidine
  • Table 2 shows the simulation results of PK parameters. It was simulated that the C max of tipepidine when the CYP2D6 inhibitor and tipepidine were combined was increased by a maximum of 3.1 times, the AUC was increased by a maximum of 9.4 times, and the t 1/2 was increased by a maximum of 4.0 times as compared with the case of administration of tipepidine alone. It was shown that the combination of tipepidine with a CYP2D6 inhibitor can be expected to increase plasma exposure and half-life of tipepidine.
  • Example 4 Evaluation of Tipepidine Blood Kinetics of Tipepidine Alone in PXB Mice and in Combination with CYP2D6 Inhibitor Quinidine and Tipepidine.
  • PXB mice which are human hepatocyte transplanted mice. It is known that there are species differences in drug metabolism in general, and the contribution ratio of various CYPs to metabolism is also different. Therefore, PXB mice transplanted with human hepatocytes are used instead of using wild-type mice or rats. It is considered that the use of is more suitable for verifying the combined effect of CYP2D6 inhibitors in humans.
  • tipepidine hibenzate alone was orally administered to male PXB mice (18-20 weeks old), which are human liver transplantation mice, or 100 mg / kg of CYP2D6 inhibitor quinidine was orally administered, and 2 hours after that, tipepidine hibenzic acid was administered. 20 mg / kg of salt was orally administered. Blood was collected at 15 minutes, 30 minutes, 1 hour, 3 hours, 6 hours, and 24 hours after administration of tipepidine hibenzate. Plasma was obtained from the collected blood, the plasma tipepidine concentration was measured by LC-MS, and the concentration transition was subjected to non-compartmental analysis using Phoenix WinNonlin (Certara), and C max , AUC, t 1 / as PK parameters. 2 was calculated.
  • tipepidine By co-administering tipepidine with CYP2D6 inhibitor quinidine, it was clarified that the C max of tipepidine was increased by 1.5 times, AUC was increased by 3.2 times, and t 1/2 was increased by 3.0 times. It has been shown that increased plasma exposure and increased half-life are expected.
  • the rate of increase shown in this Example was lower than the rate of increase simulated in Example 3, but this was due to the fact that the liver of PXB mouse was not completely humanized and the residual hepatocytes of the mouse were It is presumed that the metabolism of the drug was not affected by chidinin.
  • Example 5 Evaluation of Tipepidine Blood Kinetics of Tipepidine Alone and CYP Inhibitor 1-ABT in Combination with Tipepidine in SD Rats
  • Example 4 by combining tipepidine with CYP2D6 inhibitor quinidine in PXB mice, increase and halve in plasma exposure of tipepidine Although an increase in the period was confirmed, PXB mice are not suitable for pharmacological evaluation. Therefore, an oral administration test of tipepidine using SD rats was carried out for the purpose of confirming the exposure-increasing effect of the combined use of CYP inhibitors in rats suitable for pharmacological evaluation.
  • 1-ABT that inhibits all CYPs was used instead of the CYP2D6 inhibitor.
  • a solvent or CYP inhibitor 1-ABT 100 mg / kg was orally administered to SD male rats (7 weeks old). Two hours after the administration of the solvent or 1-ABT, 20 mg / kg of tipepidine hibenzate was orally administered. Blood was collected 30 minutes, 1 hour, 2 hours, 4 hours, and 6 hours after administration.
  • Plasma was obtained from the collected blood, the plasma tipepidine concentration was measured by LC-MS, and the concentration transition was subjected to non-compartmental analysis using Phoenix WinNonlin (Certara), and C max , AUC, t 1 / as PK parameters. 2 was calculated.
  • PK parameters are shown in Table 4. It was revealed that coadministration of tipepidine with 1-ABT increased C max 16-fold, AUC 93-fold, and t 1/2 12-fold.
  • Example 6 Examination of central side effects of tipepidine The evaluation of locomotor activity in a new environment is an evaluation system generally used for evaluating side effects of drugs acting on the central nervous system. Thirty-two SD male rats (7 weeks old) were intraperitoneally administered with a solvent or tipepidine hibenzate 25 mg / kg or 80 mg / kg. From 30 minutes after the administration, the spontaneous locomotor activity of the rats was measured for 90 minutes by a SUPERMEX apparatus (Muromachi Kikai).
  • Example 7 Antidepressant-like Action of Tipepidine Using Forced Swim Test on Wistar Rats
  • the forced swim test is widely used as a behavioral test to evaluate depression-like behavior of rats and mice, including screening for antidepressant drugs.
  • Thirty-six Wistar male rats (7 weeks old) were placed in the swimming training room on the day before the test to get accustomed to the environment. Then, the animal was put into a water tank filled with tap water at a water temperature of 25 ⁇ 1 degree, and a forced swimming training test was conducted for 15 minutes.
  • the antidepressant drug imipramine 10 mg / kg was subcutaneously administered before swimming training. After swimming, the animals were removed from the aquarium and water droplets were wiped off and returned to their cages.
  • the body weight was measured and brought into a swimming training room to get accustomed to the environment.
  • imipramine was subcutaneously administered 1 hour and 5 hours before the start of the test.
  • tipepidine administration group tipepidine hibenzate 8 mg / kg or 80 mg / kg was intraperitoneally administered 30 minutes before the start of the test.
  • Internal administration was performed. The animal was placed in a water tank filled with tap water at a water temperature of 25 ⁇ 1 ° C., a forced swimming training test was conducted for 5 minutes, and a video camera was photographed from the front. The immobility time was measured visually after blinding the moving image.
  • Example 8 Examination of central side effects of tipepidine, or a combination of tipepidine and a CYP inhibitor, SD male rats (7 weeks old) were forcibly orally administered with a solvent or the CYP inhibitor 1-ABT 100 mg / kg.
  • the vehicle-administered rat group was defined as the solvent-administered group
  • the 1-ABT-administered rat group was defined as the 1-ABT-administered group.
  • Two hours after the administration of the solvent or 1-ABT in each group the solvent or the tipepidine hibenz was 20 mg / kg of the acid salt was administered, and 20 mg / kg of tipepidine hibenzate was forcibly orally administered to the 1-ABT administration group. From 30 minutes after administration of 20 mg / kg of the solvent or tipepidine hibenzate, the spontaneous locomotor activity of the rat was measured by SUPERMEX apparatus (Muromachi Kikai) for 90 minutes.
  • Example 9 Antidepressant-like action of tipepidine by tipepidine or a combination of tipepidine and a CYP inhibitor Wistar male rats (9 weeks old) were placed in a swimming training room on the day before the test to acclimate to the environment. Then, the animal was put into a water tank filled with tap water at a water temperature of 25 ⁇ 1 degree, and a forced swimming training test was conducted for 15 minutes. After swimming, the animals were removed from the aquarium and water droplets were wiped off and returned to their cages. On the day of the test, the body weight was measured and brought into a swimming training room to get accustomed to the environment. Vehicle or CYP inhibitor 1-ABT 100 mg / kg was administered by oral gavage.
  • the solvent-administered rat group was defined as the solvent-administered group, and the 1-ABT-administered rat group was defined as the 1-ABT-administered group.
  • the solvent or tipepidine 20 mg / kg was administered as the 1-ABT-administered group.
  • tipepidine 20 mg / kg was forcibly orally administered.
  • the animal was placed in a tank filled with tap water at a water temperature of 25 ⁇ 1 ° C., and one hour after the administration of tipepidine hibenzate, or four hours later, a forced swimming training test was conducted for 5 minutes. , Taken from the front with a video camera. The immobility time was measured visually after the video was blinded.
  • Example 8 it was shown that central side effects were not observed by the combined administration of tipepidine hibenzate and a CYP inhibitor (FIG. 7). From these results, in the combined administration of tipepidine hibenzate and CYP inhibitor, compared to the administration of tipepidine hibenzate alone, central side effects are not expressed and a strong antidepressant-like action can be maintained for a long time. It became clear. In addition, since it showed a strong antidepressant effect 1 hour after administration, it was clarified that it has immediate effect.
  • Example 10 Awakening Effect of Tipepidine on Wistar Male Rats Wistar male rats (9 weeks old) were bred in a dark room with a 12-hour cycle. Rats were anesthetized by intraperitoneal administration of pentobarbital 50 mg / kg, and EEG Electrode WIRES was transplanted to the skull for measuring EEG, and EMG Electrode WIRES was transplanted around the back neck for EMG Recording. After a one week recovery period, EEG / EMG recordings were measured using Ponemah (Data Sciences International, Inc.). The Sleep stage software (KISSEI COMTEC CO., LTD) was used for the determination of the sleep stage. On the day of the test, tipepidine hibenzate 8 mg / kg and 80 mg / kg were intraperitoneally administered 30 minutes before the start of the test, and the sleep state for 4 hours thereafter was determined by software.
  • KISSEI COMTEC CO., LTD The Sleep stage software
  • FIGS. 10A, 10B, 11A and 11B The results are shown in FIGS. 10A, 10B, 11A and 11B.
  • the administration of tipepidine hibenzate 8 mg / kg and 80 mg / kg maintained the wake time as compared with the vehicle administration group. Furthermore, shortening of REM sleep time was observed at 8 mg / kg and 80 mg / kg administration. That is, it was revealed that tipepidine has a strong wakefulness effect.
  • Example 11 Effect of tipepidine for improving chronic fatigue syndrome on SD rats SD rats were bred for 3 days in a breeding box in which water kept at 23 degrees was placed at a height of 1.5 cm to prepare a model of chronic fatigue syndrome associated with sleep deprivation. .. The rats were subcutaneously administered with a solvent and tipepidine citrate at a dose of 1 mg / kg or 5 mg / kg, and 60 minutes after that, a forced swimming test was performed.
  • Example 12 Awakening Effect of Tipepidine on Wistar Rats by Tipepidine and a Combination of Tipepidine and CYP Inhibitor Wistar male rats (9 weeks old) were bred in a light-dark room with a 12-hour cycle. Rats are anesthetized by intraperitoneal administration of pentobarbital 50 mg / kg, and EEG Electrode WIRES is transplanted to the skull to measure EEG, and EMG Electrode WIRES is transplanted around the back neck for EMG Recording. After a one week recovery period, EEG / EMG recordings were measured using Ponemah (Data Sciences International, Inc.). The Sleep stage software (KISSEI COMTEC CO., LTD) was used for the determination of the sleep stage.
  • the day before the test 100 mg / kg of vehicle or CYP inhibitor 1-ABT was orally administered by gavage.
  • the vehicle-administered rat group was defined as the solvent-previous day administration group
  • the 1-ABT-administered rat group was defined as the 1-ABT previous-day administration group
  • the solvent-previous day administration group and the 1-ABT previous-day administration group were tested with solvent or tipepidine 40 mg / kg Gavage was administered 15 minutes before the start, and the sleep state for 6 hours thereafter was determined by software.
  • the results are shown in Figures 13A, 13B, 14A and 14B.
  • tipepidine hibenzate 40 mg / kg single administration group significant maintenance of wakefulness and shortening of REM sleep time were not observed as compared with the vehicle administration group.
  • the CYP inhibitor 1-ABT 100 mg / kg and tipepidine hibenzate 40 mg / kg administration groups significant awakening time maintenance and shortening of REM sleep time were observed as compared with 1-ABT 100 mg / kg and vehicle administration groups. Admitted. From these results, it was revealed that the combined administration of tipepidine hibenzate and the CYP inhibitor maintains a strong wakefulness for a long time, as compared with the administration of tipepidine hibenzate alone.
  • Example 13 Amelioration of chronic fatigue syndrome on SD rats by tipepidine or a combination of tipepidine and a CYP inhibitor.
  • SD rats were bred for 3 days in a breeding box with water kept at 23 degrees and a height of 1.5 cm. Create a chronic fatigue syndrome model.
  • vehicle or CYP inhibitor was administered by oral gavage 1 hour before the administration of tipepidine or a pharmaceutically acceptable salt thereof, and subcutaneous administration of tipepidine or a pharmaceutically acceptable salt thereof was conducted 1 hour before the start of the test. To do. A forced swimming test is carried out 60 minutes later.
  • Example 14 Awakening effect and cataplexic inhibitory effect of tipepidine on narcolepsy mouse Intraperitoneal or subcutaneous administration of solvent and tipepidine or a pharmaceutically acceptable salt thereof to narcolepsy mouse, and then videographed electroencephalogram, electromyogram and behavior for several hours. To do. Awakening time is calculated from EEG, and the frequency of cataplexy is measured from EEG electromyography and video recording.
  • Example 15 Awakening action and cataplexy inhibitory action on narcolepsy mouse by tipepidine or a combination of tipepidine and CYP inhibitor , a solvent or CYP inhibitor is forcibly orally administered 1 hour before administration of a solvent or a CYP inhibitor to tipepidine or a pharmaceutically acceptable salt thereof.
  • the oral administration of tipepidine or a pharmaceutically acceptable salt thereof is performed by oral gavage. After that, for several hours, videographs of EEG, EMG, and behavior will be taken. Awakening time is calculated from EEG, and the frequency of cataplexy is measured from EEG electromyography and video recording.
  • Example 16 Evaluation of wakefulness in narcolepsy patients by combined use of tipepidine and CYP2D6 inhibitors
  • Narcolepsy patients exhibit excessive daytime sleepiness, emotional weakness, paralysis of sleep, etc., and the prevalence in Japan is reported to be about 0.16%. ing. Although modafinil is effective against excessive daytime sleepiness, it has been reported that about 20% of patients do not respond, and further drugs are required.
  • Patients with narcolepsy are dosed with CYP2D6 inhibitor (1-300 mg) and 1-3 times daily with tipepidine or a pharmaceutically acceptable salt thereof (20-200 mg). Thereafter, a sleep maintenance test for 4 sections and 20 minutes is performed every 2 hours, and sleep latency until sleep is measured.
  • Example 17 Evaluation of emotional weakness seizures in narcolepsy patients by combination use of tipepidine and CYP2D6 inhibitors It is known that narcolepsy patients suffer from emotional weakness seizures due to excessive emotional stimulation such as emotions. Modafinil is reportedly ineffective for emotional weakness attacks. Further, gamma-hydroxybutyric acid (GBH) is used in the United States for the treatment of emotional weakness attacks in patients with narcolepsy, but it requires two doses at night, which poses a major problem in compliance. CYP2D6 inhibitor (1-300mg) and tipepidine or its pharmaceutically acceptable salt (20-200mg) were administered to patients with narcolepsy for 14 days, and the number of emotional weakness attacks for 14 days was measured. To do.
  • GSH gamma-hydroxybutyric acid
  • Example 18 Evaluation of wakefulness in sleep apnea patients by combined use of tipepidine and CYP2D6 inhibitors Sleep apnea patients become hypopnea at night, and sleep at night is subdivided so that they cannot sleep deeply and are strong in the daytime of the next day. Drowsiness, malaise, and poor concentration are required.
  • a treatment method there is used a method of improving a hypopnea state by sending pressured air using nasal continuous positive pressure breathing therapy (referred to as CPAP method). When no improvement is observed by the CPAP method, modafinil or the like suppresses excessive daytime sleepiness, but it is reported that its action is not sufficient.
  • a sleep apnea patient is administered a CYP2D6 inhibitor (1-200 mg), and is further administered 1-3 times daily with tipepidine or a pharmaceutically acceptable salt thereof (20-200 mg). Then, for 20 minutes, a 4-section sleep maintenance test is performed every 2 hours to measure sleep latency.
  • Example 19 Evaluation of wakefulness effect in Parkinson's disease patients by combined use of tipepidine and CYP2D6 inhibitors It is said that about 80% of Parkinson's disease patients have sleep disorders, which is the most frequent non-motor symptom. Particularly in patients with Parkinson's disease, strong drowsiness is observed in the daytime, which is known as one of the factors that significantly reduce the quality of life of patients. Although off-label for excessive daytime sleepiness, modafinil is recommended for use. However, the efficacy of modafinil has not been fully verified.
  • CYP2D6 inhibitor (1-300 mg) is administered to patients with Parkinson's disease, and further tipepidine or a pharmaceutically acceptable salt thereof (20-200 mg) is administered 1 to 3 times a day. Then, for 20 minutes, a 4-section sleep maintenance test is performed every 2 hours to measure sleep latency.
  • Example 20 Obsessive-compulsive effect of tipepidine for mice using the marble burying test. A cage is covered with bedding, and 20 marbles are placed on each cage. The mice are intraperitoneally or subcutaneously administered with a solvent and tipepidine or a pharmaceutically acceptable salt thereof. After administration, the mouse is placed in a cage, allowed to move freely for 15 minutes, and the number of filled glass beads is counted visually.
  • Example 21 Obsessive-compulsive amelioration effect on SD rats by tipepidine or a combination of tipepidine and a CYP inhibitor using a glass bead masking test.
  • a cage is covered with bedding, and 20 glass beads are placed on each cage.
  • the vehicle or the CYP inhibitor is orally administered by gavage to the mouse, and 1 hour after that, tipepidine or a pharmaceutically acceptable salt thereof is orally administered by gavage. After administration, the mouse is placed in a cage, allowed to move freely for 15 minutes, and the number of filled glass beads is counted visually.
  • Example 22 Post-traumatic stress disorder improving effect of tipepidine on rats using fear conditioning test Rats are placed in a chamber, and electric shock is presented from the floor grid to perform conditioning. After a fixed time (24 hours) of conditioning, the rats are intraperitoneally or subcutaneously administered with a solvent and tipepidine or a pharmaceutically acceptable salt thereof. After administration, the rat is placed in a conditioning chamber and the rate of freezing behavior in the absence of electrical stimulation is measured.
  • Example 23 Post-traumatic stress disorder ameliorative effect on SD rats by tipepidine or a combination of tipepidine and a CYP inhibitor using a fear conditioning test. Rats are placed in a chamber, and electric shock is presented from the floor grid to perform conditioning. After a certain period of conditioning (24 hours), the rats are gavaged with solvent or CYP inhibitor, and 1 hour after that, gipipidine or a pharmaceutically acceptable salt thereof is gavaged. After administration, the rat is placed in a conditioning chamber and the rate of freezing behavior in the absence of electrical stimulation is measured.
  • the present invention can be used in the fields of pharmaceuticals and the like, for example, in the fields of development or production of therapeutic and prophylactic agents for central diseases.

Landscapes

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

Abstract

The present invention relates to: a therapeutic agent or a prophylactic agent for a central nervous system disease, in which tipepidine or a pharmaceutically accepted salt thereof and CYP2D6 inhibitor are used in combination; or a therapeutic agent or a prophylactic agent, in which tipepidine alone is used, for Parkinson's disease, Parkinson's disease syndrome caused by the use of an antipsychotic drug, sleep disorders, chronic fatigue syndrome, and also for fatigue, compulsive eating, addiction, or fibromyalgia associated with nerve degeneration and central nervous system disorder, or emotional trauma and stressor-related disorders.

Description

チペピジンを含む中枢性疾患の治療剤Remedy for central diseases including tipepidine
 本発明は、チペピジンを含む中枢性疾患の治療剤に関する。 The present invention relates to a therapeutic agent for central diseases containing tipepidine.
 チペピジンは中枢性鎮咳薬として、日本において広く使用されている。チペピジンは1959年に承認された咳止め薬であり、小児患者に対しても一般的に使われる安全な薬剤である。チペピジンには幅広い薬理作用があり、中枢性鎮咳作用に加え、気分障害、疼痛、統合失調症、脳梗塞に伴う排尿障害、注意欠如・多動症、環境化学物質に起因する脳機能障害、アルツハイマー病、及び強迫症にも効果があることが報告されている。また、注意欠如・多動症及びうつ病に対して有効であることが小規模の臨床研究から明らかにされている(特許文献1、2、3、4、5、非特許文献1、2)。 Tipepidine is widely used in Japan as a central antitussive. Tipepidine is a cough medicine approved in 1959 and is a commonly used safe drug for pediatric patients. Tipepidine has a wide range of pharmacological actions, including central antitussive action, mood disorder, pain, schizophrenia, dysuria associated with cerebral infarction, attention deficit / hyperactivity disorder, cerebral dysfunction caused by environmental chemicals, Alzheimer's disease, It is also reported to be effective for obsessive-compulsive disorder. Further, it has been clarified from a small-scale clinical study that it is effective against attention deficit / hyperactivity disorder and depression ( Patent Documents 1, 2, 3, 4, 5, Non-Patent Documents 1 and 2).
 しかしながら、チペピジンは生体内半減期が1.8時間と非常に短く、1日3回の服薬が必要である。そのため、中枢性疾患において使用されるとき、血中濃度の維持が難しく、さらに薬効を示す脳内濃度に対して十分に到達できていないという課題があった(非特許文献3)。 However, tipepidine has a very short half-life in vivo of 1.8 hours, and it is necessary to take it three times a day. Therefore, when used in a central disease, there is a problem that it is difficult to maintain the blood concentration, and furthermore, the intracerebral concentration that exhibits a drug effect cannot be sufficiently reached (Non-patent Document 3).
 一方、チペピジンは上記の中枢性の疾患への効果は報告されているものの、睡眠障害(特に過眠)、慢性疲労症候群、せん妄、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、線維筋痛症、心的外傷後ストレス障害に対する有効性に関する報告はなかった(特許文献1、2、3、4、5)。 On the other hand, although tipepidine has been reported to have effects on the above central diseases, sleep disorders (especially hypersomnia), chronic fatigue syndrome, delirium, neurodegeneration and fatigue associated with central diseases, overeating, addiction, There has been no report on the efficacy against fibromyalgia and post-traumatic stress disorder ( Patent Documents 1, 2, 3, 4, 5).
 近年、睡眠障害は人口の20%程度罹患するCommon Diseaseであり、さらに、ナルコレプシーや特発性過眠障害においては、非常に強い眠気が昼間に生じることで、日常生活が著しく制限されることが知られている。また、近年、パーキンソン病の神経変性疾患に伴う過眠は40%程度の患者で生じると報告されており、今後高齢化を迎える先進国においては、日中の眠気により高齢患者のQuolity of Lifeの悪化につながると考えられている(非特許文献4、非特許文献5)。 In recent years, sleep disorder is a common disease that affects about 20% of the population. Furthermore, in narcolepsy and idiopathic hypersomnia, it has been known that extremely strong drowsiness occurs during the daytime, which significantly limits daily life. Has been. In recent years, it has been reported that about 40% of patients suffer from hypersomnia associated with Parkinson's neurodegenerative disease. In developed countries, which are aging in the future, due to daytime drowsiness, the quality of life of elderly patients will increase. It is considered to lead to deterioration (Non-Patent Documents 4 and 5).
 睡眠障害の治療にはこれまでモダフィニルやメチルフェニデートが使用されてきたが、モダフィニルにおいては、20%の患者がモダフィニルの有効性が認められないことが知られている。加えて、メチルフェニデートは依存症のリスクが高く、乱用の危険性がある習慣性医薬品であり、その使用が大きく制限されている。そこで、モダフィニルやメチルフェニデートに代わる新たな睡眠障害治療薬の開発が望まれている(非特許文献6)。 Modafinil and methylphenidate have been used to treat sleep disorders, but it is known that 20% of modafinil patients are not effective with modafinil. In addition, methylphenidate is an addictive drug with a high risk of addiction and a risk of abuse, and its use is severely restricted. Therefore, the development of a new therapeutic drug for sleep disorders in place of modafinil and methylphenidate is desired (Non-Patent Document 6).
 うつ病は気分障害のひとつに分類され、生涯有病率は10%以上と高い最も一般的な精神疾患のひとつである。主たる症状として、強い抑うつ気分、意欲・興味・喜びの低下、精神運動障害(焦燥・精神活動の制止)、食欲低下、不眠などを呈し、患者の生活の質(Quality of Life)を著しく低下させる。 Depression is classified as one of mood disorders, and one of the most common mental illnesses with a lifetime prevalence rate of 10% or higher. The main symptoms are strong depressive mood, decreased motivation / interest / joy, psychomotor disorders (frustration / inhibition of mental activity), decreased appetite, insomnia, etc., and significantly deteriorates the quality of life of patients. ..
 うつ病の治療には認知行動療法、薬物療法、電気痙攣療法(ECT)があり、中等度以上のうつ病においては抗うつ薬による薬物療法が積極的に用いられている。抗うつ薬には三環系抗うつ薬(TCA)、四環系抗うつ薬、選択的セロトニン再取り込み阻害薬(SSRI)、セロトニン・ノルアドレナリン再取り込み阻害薬、ノルアドレナリン作動性・選択的セロトニン作動性抗うつ薬(NaSSA)など複数の種類がある。しかしながら、これらの薬剤は抗うつ作用の発現に3週間以上要し、初期の単剤治療で寛解に至る患者は三分の一程度に留まるという問題点がある。また、最もよく使われているSSRIでは自殺企図や、攻撃性を生じるため、小児患者に対して抗うつ薬の使用は推奨されていない。そこで、即効性があり、自殺企図や攻撃性が起こらず安全であり、小児にも適用できる抗うつ薬の開発が望まれている。 The treatment of depression includes cognitive-behavioral therapy, drug therapy, and electroconvulsive therapy (ECT), and drug therapy with antidepressants is actively used for moderate to moderate depression. Antidepressants include tricyclic antidepressants (TCA), tetracyclic antidepressants, selective serotonin reuptake inhibitors (SSRI), serotonin / noradrenaline reuptake inhibitors, noradrenergic / selective serotonergic There are multiple types, including antidepressants (NaSSA). However, these drugs take 3 weeks or more for the onset of antidepressant action, and there is a problem that only about one-third of patients reach remission with the initial monotherapy. Also, the most commonly used SSRIs cause suicide attempts and aggression, so the use of antidepressants is not recommended for pediatric patients. Therefore, it is desired to develop an antidepressant drug that has immediate effects, is safe without causing suicide attempts or aggression, and is applicable to children.
 注意欠如・多動症(ADHD)は神経発達症群のひとつに分類され、学童期における発症率はICD10において3~7%と記載があり、代表的な小児疾患の一つである。主たる症状として、多動、衝動性並びに不注意などを呈し、学童期の生活を難しくさせる。また、青年期においてはうつ症や依存症などの重篤な精神疾患を併発させ、患者の生活の質(Quality of Life)を著しく低下させる。 Attention deficit / hyperactivity disorder (ADHD) is classified into one of the neurodevelopmental groups, and the incidence in school-age is 3 to 7% in ICD10, which is one of the typical pediatric diseases. The main symptoms are hyperactivity, impulsivity, and carelessness, which make life difficult in school. In addition, in adolescence, it causes severe mental illness such as depression and addiction, and significantly deteriorates the quality of life of the patient.
 注意欠如・多動症の治療には認知行動療法、薬物療法があり、多動衝動性を抑制するために、薬物療法が積極的に用いられている。薬物療法には中枢刺激薬(ドパイン・ノルアドレナリン再取り込み阻害薬)、非中枢刺激薬(ノルアドレナリン再取り込み阻害薬、α2アドレナリン作動薬)など複数の種類がある。しかしながら、中枢刺激薬に関してはADHDの症状を抑制する効果は強いが、依存性を形成する危険性があり乱用される恐れがあることが知られている。一方、非中枢性刺激薬は依存性を形成する危険性は少ないが、ADHDの症状を抑制する効果は弱いことが問題である。そこで、依存性の形成がなくADHDの症状を既存の中枢刺激薬並みに抑制できる薬剤が求められている。または、非中枢刺激薬もしくは中枢刺激薬と併用することで、ADHDの症状を緩和することも併せて求められている。 There are cognitive behavioral therapy and drug therapy for treatment of attention deficit / hyperactivity disorder, and drug therapy is actively used to suppress hyperkinetic impulsivity. There are several types of drug therapy, including central stimulants (dopain / noradrenaline reuptake inhibitors) and non-central stimulants (noradrenaline reuptake inhibitors, α2 adrenergic agonists). However, it is known that the central stimulants have a strong effect of suppressing the symptoms of ADHD, but have a risk of forming dependence and may be abused. On the other hand, a non-central stimulant has a low risk of forming dependence, but a problem is that the effect of suppressing the symptoms of ADHD is weak. Therefore, there is a demand for a drug that can suppress ADHD symptoms to the same level as existing central stimulants without forming dependence. Alternatively, it is also required to alleviate the symptoms of ADHD by using it together with a non-central stimulant or a central stimulant.
特開2009-227631JP-A-2009-227631 特開2011-246446Japanese Patent Laid-Open No. 2011-246446 特開2012-62272JP2012-62272A 特開2013-63958Japanese Patent Laid-Open No. 2013-63958 特開2017-36242JP, 2017-36242, A
 本発明が解決しようとする課題は、チペピジンによる中枢性疾患を治療または予防するための方法および医薬を提供することにある。 The problem to be solved by the present invention is to provide a method and a medicine for treating or preventing central diseases caused by tipepidine.
 本発明は、チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬とを併用することを特徴とする、中枢性疾患の治療剤または予防剤、中枢性疾患を治療または予防するための方法、医薬組成物およびキットを提供する。また、本発明は、チペピジンまたはその製薬学的に許容される塩含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、線維筋痛症、または心的外傷およびストレス因関連障害群の治療剤または予防剤、治療または予防するための方法、医薬組成物およびキットを提供する。 The present invention relates to a therapeutic or prophylactic agent for central diseases, a method for treating or preventing central diseases, characterized in that tipepidine or a pharmaceutically acceptable salt thereof is used in combination with a CYP2D6 inhibitor. , Pharmaceutical compositions and kits. The present invention also includes tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotic drugs, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating. The present invention provides a therapeutic or prophylactic agent for addiction, fibromyalgia, or a group of trauma and stress-related disorders, a method for treating or preventing, a pharmaceutical composition and a kit.
 すなわち、本発明は、以下のものに関する。 That is, the present invention relates to the following.
項1.
 CYP2D6阻害薬と併用される、チペピジンまたはその製薬学的に許容される塩を含む、中枢性疾患を治療または予防するための医薬組成物。
Item 1.
A pharmaceutical composition for treating or preventing a central disease, which comprises tipepidine or a pharmaceutically acceptable salt thereof in combination with a CYP2D6 inhibitor.
項2.
 チペピジンまたはその製薬学的に許容される塩と併用される、CYP2D6阻害薬を含む、中枢性疾患を治療または予防するための医薬組成物。
Item 2.
A pharmaceutical composition for treating or preventing a central disease, comprising a CYP2D6 inhibitor, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
項3.
 チペピジンまたはその製薬学的に許容される塩およびCYP2D6阻害薬を含む、中枢性疾患を治療または予防するための医薬組成物。
Item 3.
A pharmaceutical composition for treating or preventing a central disease, comprising tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
項4.
 CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブ、デュロキセチン、ミラベグロン、セレコキシブ、エスシタロプラム、モクロベミド、シメチジン、フルボキサミン、アビラテロン、アミオダロン、クロバザム、コビシスタット、デスベンラファキシン、ラベタロール、ロルカセリン、リトナビル、セルトラリン、ベムラフェニブ、およびデラムシクランからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、上記項1~3のいずれか一項に記載の医薬組成物。
Item 4.
CYP2D6 inhibitors are quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, duloxetine, mirabegron, celecoxib, escitalopram, moclobelatil, clavitone, fluvoxamine, abiraterabalone, mirabedalone, amiodarone, fluvoxamine, abiraterabalone, amiodalone, amiodala. 4. The pharmaceutical composition according to any one of the above items 1 to 3, which is one or more drugs selected from the group consisting of, ritonavir, sertraline, vemurafenib, and deramciclane, or a pharmaceutically acceptable salt thereof. ..
項5.
 CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブ、デュロキセチン、ミラベグロン、セレコキシブ、エスシタロプラム、モクロベミド、シメチジン、およびフルボキサミンからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、上記項1~3のいずれか一項に記載の医薬組成物。
Item 5.
The CYP2D6 inhibitor is one or more pharmaceutical agents selected from the group consisting of quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, duloxetine, mirabegron, celecoxib, escitalopram, moclobemide, cimetidine, and fluvoxamine; Item 4. The pharmaceutical composition according to any one of Items 1 to 3, wherein the pharmaceutical composition is a salt acceptable for.
項6.
 CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、およびダコミチニブからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、上記項1~3のいずれか一項に記載の医薬組成物。
Item 6.
1 to 3 above, wherein the CYP2D6 inhibitor is one or more drugs selected from the group consisting of quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, and dacomitinib, or a pharmaceutically acceptable salt thereof. The pharmaceutical composition according to any one of claims.
項7.
 CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、およびフルオキセチンからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、上記項1~3のいずれか一項に記載の医薬組成物。
Item 7.
Item 4. The CYP2D6 inhibitor is one or more drugs selected from the group consisting of quinidine, paroxetine, bupropion, and fluoxetine, or a pharmaceutically acceptable salt thereof, according to any one of items 1 to 3 above. Pharmaceutical composition.
項8.
 CYP2D6阻害薬が、キニジン、またはその製薬学的に許容される塩である、上記項1~3のいずれか一項に記載の医薬組成物。
Item 8.
Item 4. The pharmaceutical composition according to any one of Items 1 to 3, wherein the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof.
項9.
 中枢性疾患が、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 9.
Central diseases are Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central diseases, overeating, addiction, or fibromyalgia, the above-mentioned item The pharmaceutical composition according to any one of 1 to 8.
項10.
 中枢性疾患が、睡眠障害である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 10.
Item 9. The pharmaceutical composition according to any one of Items 1 to 8, wherein the central disease is a sleep disorder.
項11.
 中枢性疾患が、過眠、特発性過眠障害、またはナルコレプシーである、上記項1~8および10のいずれか一項に記載の医薬組成物。
Item 11.
11. The pharmaceutical composition according to any one of items 1 to 8 and 10, wherein the central disease is hypersomnia, idiopathic hypersomnia disorder, or narcolepsy.
項12.
 中枢性疾患が、抑うつ障害群である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 12.
9. The pharmaceutical composition according to any one of items 1 to 8, wherein the central disease is a depressive disorder group.
項13.
 中枢性疾患が、不安性の苦痛を伴う抑うつ障害群、混合性の特徴を伴う抑うつ障害群、メランコリアの特徴を伴う抑うつ障害群、気分に一致する精神病性の特徴を伴う抑うつ障害群、気分に一致しない精神病性の特徴を伴う抑うつ障害群、緊張病を伴う抑うつ障害群、季節型抑うつ障害群、重篤気分調整症、うつ病、持続性抑うつ障害、月経前不快気分障害、または物質・医薬品誘発性抑うつ障害である、上記項1~8および12のいずれか一項に記載の医薬組成物。
Item 13.
Central disorders include anxiety-related depressive disorders, depressive disorders with mixed features, depressive disorders with melancholic features, depressive disorders with mood-matching psychotic features, mood Depressive disorders with inconsistent psychotic features, depressive disorders with catatonic disorders, seasonal depressive disorders, severe dysregulation, depression, persistent depressive disorder, premenstrual dysphoric disorder, or substances / medicines 13. The pharmaceutical composition according to any one of the above items 1 to 8 and 12, which is an induced depressive disorder.
項14.
 中枢性疾患が、小児における抑うつ障害群である、上記項1~8および12のいずれか一項に記載の医薬組成物。
Item 14.
13. The pharmaceutical composition according to any one of items 1 to 8 and 12, wherein the central disease is a group of depressive disorders in children.
項15.
 中枢性疾患が、小児における不安性の苦痛を伴う抑うつ障害群、小児における混合性の特徴を伴う抑うつ障害群、小児におけるメランコリアの特徴を伴う抑うつ障害群、小児における気分に一致する精神病性の特徴を伴う抑うつ障害群、小児における気分に一致しない精神病性の特徴を伴う抑うつ障害群、小児における緊張病を伴う抑うつ障害群、小児における季節型抑うつ障害群、小児における重篤気分調整症、小児におけるうつ病、小児における持続性抑うつ障害、小児における月経前不快気分障害、または小児における物質・医薬品誘発性抑うつ障害である、上記項1~8および14のいずれか一項に記載の医薬組成物。
Item 15.
Central disorders are anxiety-affected depressive disorders in children, depressive disorders with mixed features in children, depressive disorders with melancholic features in children, psychotic features that match mood in children Depressive disorder group with illness, depressive disorder group with psychotic features inconsistent with mood in children, depressive disorder group with catatonic disorders in children, seasonal depressive disorder group in children, severe mood regulation in children, in children 15. The pharmaceutical composition according to any one of the above items 1 to 8 and 14, which is depression, persistent depressive disorder in children, premenstrual dysphoric disorder in children, or substance / medicine-induced depressive disorder in children.
項16.
 中枢性疾患が、不安症群である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 16.
9. The pharmaceutical composition according to any one of items 1 to 8 above, wherein the central disease is an anxiety group.
項17.
 中枢性疾患が、分離不安症、限局性恐怖症、社交不安症、パニック症、または物質・医薬品誘発性不安症である、上記項1~8および16のいずれか一項に記載の医薬組成物。
Item 17.
17. The pharmaceutical composition according to any one of the above items 1 to 8 and 16, wherein the central disease is separation anxiety disorder, localized phobia, social anxiety disorder, panic disorder, or substance / drug-induced anxiety disorder. ..
項18.
 中枢性疾患が、小児における不安症群である、上記項1~8および16のいずれか一項に記載の医薬組成物。
Item 18.
17. The pharmaceutical composition according to any one of items 1 to 8 and 16 above, wherein the central disease is an anxiety group in children.
項19.
 中枢性疾患が、小児における分離不安症、小児における限局性恐怖症、小児における社交不安症、小児におけるパニック症、または小児における物質・医薬品誘発性不安症である、上記項1~8および18のいずれか一項に記載の医薬組成物。
Item 19.
The central disease is separation anxiety in children, localized phobia in children, social anxiety in children, panic disorder in children, or substance / drug-induced anxiety in children. The pharmaceutical composition according to any one of claims.
項20.
 中枢性疾患が、神経発達症群である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 20.
Item 9. The pharmaceutical composition according to any one of Items 1 to 8, wherein the central disease is a neurodevelopmental group.
項21.
 中枢性疾患が、自閉症スペクトラム症、注意欠如・多動症、限局性学習症、知的能力障害、言語症、語音症、小児期発症流暢症、社会的(語用論的)コミュニケーション症、発達性協調運動症、またはチック症群である、上記項1~8および20のいずれか一項に記載の医薬組成物。
Item 21.
Central diseases include autism spectrum disorder, attention deficit / hyperactivity disorder, localized learning disorder, intellectual disability, speech disorders, speech disorders, childhood-onset fluency, social (pragmatic) communication disorders, development 21. The pharmaceutical composition according to any one of items 1 to 8 and 20 above, which is a group having sexual dyskinesia or tic disorder.
項22.
 中枢性疾患が、強迫症および関連症群である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 22.
9. The pharmaceutical composition according to any one of items 1 to 8 above, wherein the central disease is a group of obsessive-compulsive disorder and related disorders.
項23.
 中枢性疾患が、強迫症、醜形恐怖症、ためこみ症、抜毛症、または皮膚むしり症である、上記項1~8および22のいずれか一項に記載の医薬組成物。
Item 23.
23. The pharmaceutical composition according to any one of the above items 1 to 8 and 22, wherein the central disease is obsessive-compulsive disorder, dysphoric phobia, dentosis, hair loss, or plucking skin.
項24.
 中枢性疾患が、心的外傷およびストレス因関連障害群である、上記項1~8のいずれか一項に記載の医薬組成物。
Item 24.
9. The pharmaceutical composition according to any one of items 1 to 8, wherein the central disease is a group of disorders related to trauma and stress factors.
項25.
 中枢性疾患が、心的外傷後ストレス障害、反応性アタッチメント障害、脱抑制型対人交流障害、急性ストレス障害、または適応障害である、上記項1~8および24のいずれか一項に記載の医薬組成物。
Item 25.
25. The medicine according to any one of the above items 1 to 8 and 24, wherein the central disease is post-traumatic stress disorder, reactive attachment disorder, disinhibition-type interpersonal interaction disorder, acute stress disorder, or adaptation disorder. Composition.
項26.
 チペピジンまたはその製薬学的に許容される塩の投与量が一日当たり2mg~2000mgである、上記項1~25のいずれか一項に記載の医薬組成物。
Item 26.
The pharmaceutical composition according to any one of the above items 1 to 25, wherein the dose of tipepidine or a pharmaceutically acceptable salt thereof is 2 mg to 2000 mg per day.
項27.
 CYP2D6阻害薬の投与量が一日当たり1mg~1000mgである、上記項1~26のいずれか一項に記載の医薬組成物。
Item 27.
27. The pharmaceutical composition according to any one of items 1 to 26 above, wherein the dose of the CYP2D6 inhibitor is 1 mg to 1000 mg per day.
項28.
 治療上の有効量のチペピジンまたはその製薬学的に許容される塩と治療上の有効量のCYP2D6阻害薬を哺乳動物に投与することを含む、中枢性疾患を治療または予防するための方法。
Item 28.
A method for treating or preventing a central disease comprising administering to a mammal a therapeutically effective amount of tipepidine or a pharmaceutically acceptable salt thereof and a therapeutically effective amount of a CYP2D6 inhibitor.
項29.
 CYP2D6阻害薬と併用される、中枢性疾患の治療または予防における使用のための、チペピジンまたはその製薬学的に許容される塩。
Item 29.
Tipepidine or a pharmaceutically acceptable salt thereof for use in the treatment or prevention of central diseases in combination with a CYP2D6 inhibitor.
項30.
 チペピジンまたはその製薬学的に許容される塩と併用される、中枢性疾患の治療または予防における使用のための、CYP2D6阻害薬。
Item 30.
A CYP2D6 inhibitor for use in the treatment or prevention of central diseases, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
項31.
 CYP2D6阻害薬と併用される、中枢性疾患を治療または予防するための医薬の製造のための、チペピジンまたはその製薬学的に許容される塩の使用。
Item 31.
Use of tipepidine or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing central diseases, which is used in combination with a CYP2D6 inhibitor.
項32.
 チペピジンまたはその製薬学的に許容される塩と併用される、中枢性疾患を治療または予防するための医薬の製造のための、CYP2D6阻害薬の使用。
Item 32.
Use of a CYP2D6 inhibitor for the manufacture of a medicament for treating or preventing central diseases, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
項33.
 中枢性疾患を治療または予防するための医薬の製造のための、チペピジンまたはその製薬学的に許容される塩およびCYP2D6阻害薬の使用。
Item 33.
Use of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor for the manufacture of a medicament for treating or preventing central diseases.
項34.
 チペピジンまたはその製薬学的に許容される塩と、CYP2D6阻害薬を含む、中枢性疾患の治療または予防のためのキット。
Item 34.
A kit for the treatment or prevention of central diseases, which comprises tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
項35.
 チペピジンまたはその製薬学的に許容される塩を含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、線維筋痛症、または心的外傷およびストレス因関連障害群の治療剤または予防剤。
Item 35.
Parkinson's disease, Parkinson's syndrome due to antipsychotic use, sleep disorder, chronic fatigue syndrome, fatigue associated with neurodegeneration and central illness, overeating, addiction, fiber containing tipepidine or a pharmaceutically acceptable salt thereof A therapeutic or prophylactic agent for myalgia, or trauma and stress-related disorders.
項36.
 チペピジンまたはその製薬学的に許容される塩を含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症の治療剤または予防剤。
Item 36.
Including tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorder, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating, addiction, or A therapeutic or preventive agent for fibromyalgia.
項37.
 疾患が、睡眠障害である、上記項35または36に記載の治療剤または予防剤。
Item 37.
Item 37. The therapeutic or prophylactic agent according to Item 35 or 36, wherein the disease is a sleep disorder.
項38.
 疾患が、過眠、特発性過眠障害、またはナルコレプシーである、上記項35または36に記載の治療剤または予防剤。
Item 38.
37. The therapeutic or prophylactic agent according to the above item 35 or 36, wherein the disease is hypersomnia, idiopathic hypersomnia disorder, or narcolepsy.
項39.
 疾患が、心的外傷およびストレス因関連障害群である、上記項35または36に記載の治療剤または予防剤。
Item 39.
Item 37. The therapeutic or prophylactic agent according to Item 35 or 36 above, wherein the disease is a group of trauma and stress-related disorders.
項40.
 疾患が、心的外傷後ストレス障害、反応性アタッチメント障害、脱抑制型対人交流障害、急性ストレス障害、または適応障害である、上記項35または36に記載の治療剤または予防剤。
Item 40.
37. The therapeutic or prophylactic agent according to the above item 35 or 36, wherein the disease is post-traumatic stress disorder, reactive attachment disorder, disinhibition-type interpersonal interaction disorder, acute stress disorder, or adaptation disorder.
項41.
 疾患が、心的外傷後ストレス障害である、上記項35または36に記載の治療剤または予防剤。
Item 41.
37. The therapeutic or prophylactic agent according to the above item 35 or 36, wherein the disease is post-traumatic stress disorder.
 本発明はまた、以下のものに関する:
 チペピジンまたはその製薬学的に許容される塩を哺乳動物に投与することを含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、線維筋痛症、または心的外傷およびストレス因関連障害群を治療または予防するための方法;
 パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、線維筋痛症、または心的外傷およびストレス因関連障害群の治療または予防における使用のための、チペピジンまたはその製薬学的に許容される塩;
 パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、線維筋痛症、または心的外傷およびストレス因関連障害群を治療または予防するための医薬の製造のための、チペピジンまたはその製薬学的に許容される塩の使用;および
 疾患が、睡眠障害(特に、過眠、特発性過眠障害、またはナルコレプシー)である、これらの発明。
The invention also relates to:
Fatigue associated with Parkinson's disease, Parkinson's disease syndrome due to antipsychotic drug use, sleep disorders, chronic fatigue syndrome, neurodegeneration and central illness, including administration of tipepidine or a pharmaceutically acceptable salt thereof to a mammal , A method for treating or preventing hyperphagia, addiction, fibromyalgia, or trauma and stress-related disorders.
Parkinson's disease, Parkinson's syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central illness, overeating, addiction, fibromyalgia, or trauma and stress-related disorders Or a pharmaceutically acceptable salt thereof for use in the treatment or prophylaxis of
Parkinson's disease, Parkinson's syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central illness, overeating, addiction, fibromyalgia, or trauma and stress-related disorders Use of tipepidine or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prophylaxis; and if the disease is a sleep disorder (especially hypersomnia, idiopathic hypersomnia disorder, or narcolepsy). There are these inventions.
 本発明により、チペピジンまたはその製薬学的に許容される塩にCYP2D6阻害薬を併用させることで、生体内半減期が短かったチペピジンの薬物動態を改善し、またチペピジンの新たな中枢作用を見出したことにより、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症といった中枢性疾患の治療剤または予防剤、中枢性疾患を治療または予防するための方法、医薬組成物およびキットを提供することが可能となった。特に、本発明は睡眠障害の改善に期待できる。また、本発明により、チペピジンまたはその製薬学的に許容される塩含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症の治療剤または予防剤、治療または予防するための方法、医薬組成物およびキットも提供される。また、本発明により、チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬とを併用することによる、抑うつ障害群、不安症群、神経発達症群、強迫症および関連症群、または心的外傷およびストレス因関連障害群の治療剤もしくは予防剤、治療または予防するための方法、医薬組成物およびキットが提供される。また、本発明により、チペピジンまたはその製薬学的に許容される塩を含む、心的外傷およびストレス因関連障害群の治療剤または予防剤、治療または予防するための方法、医薬組成物およびキットも提供される。 According to the present invention, by combining tipepidine or a pharmaceutically acceptable salt thereof with a CYP2D6 inhibitor, the pharmacokinetics of tipepidine having a short half-life in vivo were improved, and a new central action of tipepidine was found. Thereby, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotics, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating, dependence, or therapeutic agents for central diseases such as fibromyalgia Alternatively, it has become possible to provide a preventive agent, a method for treating or preventing a central disease, a pharmaceutical composition and a kit. In particular, the present invention can be expected to improve sleep disorders. Further, according to the present invention, comprising tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotic drugs, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating. Also provided are therapeutic or prophylactic agents for addiction, or fibromyalgia, methods for treating or preventing, pharmaceutical compositions and kits. In addition, according to the present invention, by using tipepidine or a pharmaceutically acceptable salt thereof in combination with a CYP2D6 inhibitor, a depressive disorder group, anxiety group, neurodevelopmental group, obsessive-compulsive and related disease group, or heart. Provided are therapeutic agents or prophylactic agents for the group of trauma and stress-related disorders, methods for treating or preventing, pharmaceutical compositions and kits. Further, according to the present invention, a therapeutic or prophylactic agent for a group of disorders related to trauma and stress factors, a method, a pharmaceutical composition and a kit for treating or preventing, comprising tipepidine or a pharmaceutically acceptable salt thereof. Provided.
図1はヒト肝細胞におけるチペピジンの代謝安定性を示したものであり、CYP阻害薬である1-ABTを添加した処置群と非処置群におけるチペピジンの残存率をそれぞれ示している。横軸には反応時間、縦軸にはチペピジンの残存率を示す。FIG. 1 shows the metabolic stability of tipepidine in human hepatocytes, and shows the residual rates of tipepidine in the treated and non-treated groups to which 1-ABT, a CYP inhibitor, was added. The horizontal axis shows the reaction time, and the vertical axis shows the residual ratio of tipepidine. 図2はヒトにチペピジン単独、CYP2D6阻害薬とチペピジンを併用して経口投与した時のチペピジンの血漿中濃度推移をシミュレーションし示したものである。横軸には投与からの時間、縦軸にはチペピジンの血漿中濃度を示す。FIG. 2 shows simulated changes in plasma concentration of tipepidine when orally administered to humans alone with tipepidine or with combined use of a CYP2D6 inhibitor and tipepidine. The horizontal axis shows the time from administration, and the vertical axis shows the plasma concentration of tipepidine. 図3はヒト肝細胞移植マウス(PXBマウス)にチペピジンヒベンズ酸塩20mg/kgを単独、CYP2D6阻害薬キニジン100mg/kgとチペピジンヒベンズ酸塩20mg/kgを併用して経口投与した時のチペピジンの血漿中濃度推移を示したものである。横軸には投与からの時間、縦軸にはチペピジンの血漿中濃度を示す。FIG. 3 shows plasma of tipepidine when orally administered to human hepatocyte-transplanted mice (PXB mice) with 20 mg / kg of tipepidine hibenzate alone and with 100 mg / kg of CYP2D6 inhibitor quinidine and 20 mg / kg of tipepidine hibenzate. The figure shows the transition of medium concentration. The horizontal axis shows the time from administration, and the vertical axis shows the plasma concentration of tipepidine. 図4はSDラットにチペピジンヒベンズ酸塩20mg/kgを単独、CYP阻害薬1-ABT100mg/kgとチペピジンヒベンズ酸塩20mg/kgを併用して経口投与した時のチペピジンの血漿中濃度推移を示したものである。横軸には投与からの時間、縦軸にはチペピジンの血漿中濃度を示す。FIG. 4 shows changes in plasma concentrations of tipepidine when SD rats were orally administered with 20 mg / kg of tipepidine hibenzate alone, and combined with 20 mg / kg of CYP inhibitor 1-ABT and 100 mg / kg of tipepidine hibenzate. It is a thing. The horizontal axis shows the time from administration, and the vertical axis shows the plasma concentration of tipepidine. 図5は、SDラットに溶媒、又はチペピジンヒベンズ酸塩を腹腔内投与し(25mg/kgおよび80mg/kgの2群)、投与30分後から1.5時間の間の自発運動量の総和を表示したものである。なお、棒グラフは各処置群の平均値を示し、**はダネット検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 5 shows the total amount of locomotor activity from 30 minutes after administration to 1.5 hours after intraperitoneal administration of solvent or tipepidine hibenzate (2 groups of 25 mg / kg and 80 mg / kg) to SD rats. It was done. The bar graph shows the average value of each treatment group, and ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01. 図6は、SDラットに溶媒を腹腔内投与、イミプラミン10mg/kgを皮下投与、又はチペピジンヒベンズ酸塩(8.0mg/kgおよび80mg/kgの2群)を腹腔内投与し、その30分後に強制水泳試験を行い、ラットの無動時間(秒)を表示したものである。なお、棒グラフは各処置群の平均値を示し、**はチューキー・クレーマー検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 6 shows that SD rats were intraperitoneally administered with a solvent, imipramine 10 mg / kg was subcutaneously administered, or tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg) was intraperitoneally administered 30 minutes later. The forced swimming test was performed, and the immobility time (seconds) of the rat was displayed. The bar graph shows the average value of each treatment group, and ** means that the risk rate (P) calculated by the Tukey-Kramer test is less than 0.01. 図7は、SDラットに溶媒、チペピジンヒベンズ酸塩20mg/kg、又はチペピジンヒベンズ酸塩20mg/kgとCYP阻害薬1-ABT100mg/kgを強制経口投与し、チペピジン投与30分後から1.5時間の自発運動量の総和を表示したものである。なお、棒グラフは各処置群の平均値を示し、N.S.はチューキー・クレーマー検定によって算出した危険率(P)が0.05以上であることを意味する。FIG. 7 shows that SD rats were forcibly orally administered with vehicle, tipepidine hibenzate 20 mg / kg, or tipepidine hibenzate 20 mg / kg and CYP inhibitor 1-ABT 100 mg / kg, and 30 minutes after the administration of tipepidine, 1.5 hours. It is a display of the total sum of the spontaneous locomotor activity of. The bar graph shows the average value of each treatment group. S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more. 図8は、SDラットに溶媒、チペピジンヒベンズ酸塩20mg/kg、もしくはチペピジンヒベンズ酸塩20mg/kg及びCYP阻害薬1-ABT100mg/kgを強制経口投与し、その投与1時間後に強制水泳試験を行い、ラットの無動時間(秒)を表示したものである。なお、棒グラフは各処置群の平均値を示し、N.S.はチューキー・クレーマー検定によって算出した危険率(P)が0.05以上であること、**はチューキー・クレーマー検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 8 shows that SD rats were forcibly orally administered with solvent, tipepidine hibenzate 20 mg / kg, or tipepidine hibenzate 20 mg / kg and CYP inhibitor 1-ABT 100 mg / kg, and a forced swimming test was conducted 1 hour after the administration. , The immobility time of the rat (second) is displayed. The bar graph shows the average value of each treatment group. S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more, and ** means that the risk factor (P) calculated by the Tukey-Kramer test is less than 0.01. 図9は、SDラットに溶媒、チペピジンヒベンズ酸塩20mg/kg、もしくはチペピジンヒベンズ酸塩20mg/kg及びCYP阻害薬1-ABT100mg/kgを強制経口投与し、その投与4時間後に強制水泳試験を行い、ラットの無動時間(秒)を表示したものである。なお、棒グラフは各処置群の平均値を示し、N.S.はチューキー・クレーマー検定によって算出した危険率(P)が0.05以上であること、**はチューキー・クレーマー検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 9 shows that SD rats were forcibly orally administered with solvent, tipepidine hibenzate 20 mg / kg, or tipepidine hibenzate 20 mg / kg and CYP inhibitor 1-ABT 100 mg / kg, and a forced swimming test was conducted 4 hours after the administration. , The immobility time of the rat (second) is displayed. The bar graph shows the average value of each treatment group. S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more, and ** means that the risk factor (P) calculated by the Tukey-Kramer test is less than 0.01. 図10Aは、Wistarラットに溶媒、チペピジンヒベンズ酸塩(8.0mg/kgおよび80mg/kgの2群)を腹腔内投与し、投与後4時間の睡眠脳波の測定を行い、ラットの覚醒時間を表示したものである。FIG. 10A shows that Wistar rats were intraperitoneally administered with a solvent, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg), sleep EEG was measured 4 hours after the administration, and the awakening time of the rats was measured. It is displayed. 図10Bは、Wistarラットに溶媒、チペピジンヒベンズ酸塩(8.0mg/kgおよび80mg/kgの2群)を腹腔内投与し、投与後4時間の総覚醒時間を表示したものである。なお、棒グラフは各処置群の平均値を示し、**はダネット検定によって算出した危険率(P)が0.01未満であることを意味し、*はダネット検定によって算出した危険率(P)が0.05未満であることを意味する。FIG. 10B is a graph showing the total wakeup time 4 hours after the administration of the solvent, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg) to the Wistar rat, intraperitoneally. The bar graph shows the average value of each treatment group, ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01, and * indicates the risk rate (P) calculated by Dunnett's test. Means less than 0.05. 図11Aは、Wistarラットに溶媒、チペピジンヒベンズ酸塩(8.0mg/kgおよび80mg/kgの2群)を腹腔内投与した。投与後4時間のレム睡眠脳波の測定を行い、ラットのレム睡眠時間を表示したものである。FIG. 11A shows that Wistar rats were intraperitoneally administered with a solvent, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg). REM sleep EEG was measured 4 hours after administration, and the REM sleep time of the rat was displayed. 図11Bは、Wistarラットに溶媒、チペピジンヒベンズ酸塩(8.0mg/kgおよび80mg/kgの2群)を腹腔内投与し、投与後4時間の総レム睡眠時間を表示したものである。なお、棒グラフは各処置群の平均値を示し、**はダネット検定によって算出した危険率(P)が0.01未満であることを意味し、*はダネット検定によって算出した危険率(P)が0.05未満であることを意味する。FIG. 11B is a graph showing the total REM sleep time of 4 hours after the intraperitoneal administration of the vehicle, tipepidine hibenzate (2 groups of 8.0 mg / kg and 80 mg / kg), to Wistar rats. The bar graph shows the average value of each treatment group, ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01, and * indicates the risk rate (P) calculated by Dunnett's test. Means less than 0.05. 図12は慢性的に疲労させたSDラットに、溶媒及びチペピジンクエン酸塩1mg/kg、又は5mg/kgの用量で皮下投与を行い、投与60分後の強制水泳試験による無動時間を表示したものである。**はダネット検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 12 shows the immobility time by the forced swimming test 60 minutes after the administration, which was subcutaneously administered to a chronically fatigued SD rat at a dose of 1 mg / kg of solvent and tipepidine citrate or 5 mg / kg. It is a thing. ** means that the risk rate (P) calculated by Dunnett's test is less than 0.01. 図13Aは、試験前日にWistarラットに溶媒又はCYP阻害薬1-ABT100mg/kgを強制経口投与し、試験開始の15分前に溶媒又はチペピジンヒベンズ酸塩40mg/kgを強制経口投与し、試験開始後6時間の睡眠脳波の測定を行い、ラットの覚醒時間を表示したものである。FIG. 13A shows that on the day before the test, Wistar rats were forcibly orally administered with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the solvent or tipepidine hibenzate 40 mg / kg was orally administered, and the test was started. The sleep electroencephalogram was measured 6 hours later, and the awakening time of the rat was displayed. 図13Bは、試験前日にWistarラットに溶媒又はCYP阻害薬1-ABT100mg/kgを強制経口投与し、試験開始の15分前に溶媒又はチペピジンヒベンズ酸塩40mg/kgを強制経口投与し、試験開始後6時間の総覚醒時間を表示したものである。なお、棒グラフは各処置群の平均値を示し、N.S.はチューキー・クレーマー検定によって算出した危険率(P)が0.05以上であること、**はチューキー検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 13B shows that on the day before the test, Wistar rats were gavaged with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the vehicle or the tipepidine hibenzate 40 mg / kg was gavaged, and the test was started. It shows the total awakening time of the next 6 hours. The bar graph shows the average value of each treatment group. S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more, and ** means that the risk factor (P) calculated by the Tukey test is less than 0.01. 図14Aは、試験前日にWistarラットに溶媒又はCYP阻害薬1-ABT100mg/kgを強制経口投与し、試験開始の15分前に溶媒又はチペピジンヒベンズ酸塩40mg/kgを強制経口投与し、試験開始後6時間のレム睡眠脳波の測定を行い、ラットのレム睡眠時間を表示したものである。FIG. 14A shows that on the day before the test, Wistar rats were gavaged with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the vehicle or the tipepidine hibenzate salt 40 mg / kg was gavaged orally, and the test was started. The REM sleep electroencephalogram was measured 6 hours later, and the REM sleep time of the rat was displayed. 図14Bは、試験前日にWistarラットに溶媒又はCYP阻害薬1-ABT100mg/kgを強制経口投与し、試験開始の15分前に溶媒又はチペピジンヒベンズ酸塩40mg/kgを強制経口投与し、試験開始後6時間の総レム睡眠時間を表示したものである。なお、棒グラフは各処置群の平均値を示し、N.S.はチューキー・クレーマー検定によって算出した危険率(P)が0.05以上であること、**はチューキー検定によって算出した危険率(P)が0.01未満であることを意味する。FIG. 14B shows that on the day before the test, Wistar rats were gavaged with the solvent or the CYP inhibitor 1-ABT 100 mg / kg, and 15 minutes before the start of the test, the vehicle or the tipepidine hibenzate 40 mg / kg was gavaged, and the test was started. It is a display of the total REM sleep time of the next 6 hours. The bar graph shows the average value of each treatment group. S. Means that the risk factor (P) calculated by the Tukey-Kramer test is 0.05 or more, and ** means that the risk factor (P) calculated by the Tukey test is less than 0.01.
 以下、本発明の実施形態について詳細に説明する。当該実施形態は本発明の原理の理解を容易にするためのものであり、本発明の範囲は、下記の実施形態に限られるものではなく、当業者が以下の実施形態の構成を適宜置換した他の実施形態も、本発明の範囲に含まれる。 Hereinafter, embodiments of the present invention will be described in detail. The embodiment is for facilitating the understanding of the principle of the present invention, and the scope of the present invention is not limited to the following embodiment, and those skilled in the art appropriately replace the configuration of the following embodiment. Other embodiments are also within the scope of the invention.
 本発明の一つの態様は、チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬との併用に関する。 One aspect of the present invention relates to a combination of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
 本発明の別の態様は、チペピジンまたはその製薬学的に許容される塩を含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症の治療剤または予防剤に関する。 Another aspect of the present invention comprises tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central diseases. The present invention relates to a therapeutic or preventive agent for feeling, overeating, addiction, or fibromyalgia.
 本発明における「チペピジン」は、化学名が3-(ジチエン-2-イルメチレン)-1-メチルピペリジンであり、市販の鎮咳薬の有効成分として既に使用されている。そして、チペピジンとしては、遊離塩基体またはその製薬学的に許容される酸付加塩および/または水和物および/または溶媒和物で用いられ得る(本明細書においては、これらを総称してチペピジンまたはその製薬学的に許容される塩という)。適当な酸付加塩としては、例えば、コハク酸、臭化水素酸、酢酸、フマル酸、マレイン酸、メタンスルホン酸、乳酸、リン酸、塩酸、硫酸、酒石酸、クエン酸およびヒベンズ酸から選択される酸の塩が挙げられ、好ましくは、ヒベンズ酸またはクエン酸の塩が挙げられる。 “Tipepidine” in the present invention has a chemical name of 3- (dithien-2-ylmethylene) -1-methylpiperidine and has already been used as an active ingredient of a commercially available antitussive drug. And, as tipepidine, it can be used in a free base form or a pharmaceutically acceptable acid addition salt and / or hydrate and / or solvate thereof (in the present specification, these are collectively referred to as tipepidine Or its pharmaceutically acceptable salt). Suitable acid addition salts are, for example, selected from succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulfonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulfuric acid, tartaric acid, citric acid and hibenzic acid. Examples thereof include salts of acids, preferably salts of hibenzic acid or citric acid.
 薬物代謝に関する酵素の内、主要な役割を示す酵素としてシトクロムP450(以下、CYPという)が知られている。CYPには多くの分子種が知られているが、主要な分子種はCYP1A2、CYP2B6、CYP2C8、CYP2C9、CYP2C19、CYP2D6及びCYP3A4/5である。CYP2D6阻害薬は、CYP2D6の酵素活性を抑制する作用を有することから、CYP2D6によって代謝される薬剤(以下、CYP2D6基質薬という)と併用した場合にCYP2D6基質薬の血中曝露を増加させる。 Among the enzymes involved in drug metabolism, cytochrome P450 (hereinafter referred to as CYP) is known as an enzyme that plays a major role. Although many molecular species are known for CYP, the major molecular species are CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A4 / 5. Since the CYP2D6 inhibitor has an action of suppressing the enzymatic activity of CYP2D6, it increases blood exposure of the CYP2D6 substrate drug when used in combination with a drug metabolized by CYP2D6 (hereinafter referred to as CYP2D6 substrate drug).
 本発明における「CYP2D6阻害薬」とは、臨床薬物相互作用試験において、相互作用を受けやすいCYP2D6基質薬の血漿中曝露(AUC)を1.25倍以上、好ましくは2倍以上、より好ましくは5倍以上に上昇させる阻害能を有する薬剤を意味する。本発明における「CYP2D6阻害薬」として、具体的には、平成26年7月8日発行の「医薬品開発と適正な情報提供のための薬物相互作用ガイドライン(最終案)」または2016年9月26日にアメリカ食品医薬品局(FDA)が発行したDrug Development and Drug Interactions: Table of Substrates, Inhibitors and InducersにおけるTable 3-2: Examples of clinical inhibitors for P450-mediated metabolisms (for concomitant use clinical DDI studies and/or drug labeling)においてCYP2D6の強い阻害薬として分類されているキニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブ、中程度の阻害薬として分類されているデュロキセチン、ミラベグロン、セレコキシブ、エスシタロプラム、モクロベミド、シメチジン、フルボキサミン、弱い阻害薬として分類されているアビラテロン、アミオダロン、クロバザム、コビシスタット、デスベンラファキシン、ラベタロール、ロルカセリン、リトナビル、セルトラリン、ベムラフェニブ、デラムシクランまたはそれらの製薬学的に許容される塩が挙げられる。本発明における「CYP2D6阻害薬」としては、これらの薬剤の任意の2つ以上を組み合わせて用いてもよい。これらの薬剤は、主作用としては別の作用を有するが、CYP2D6阻害作用を併せ持つため、本明細書においては「CYP2D6阻害薬」としている。これらは、遊離塩基体またはその薬学上許容される酸付加塩および/または水和物および/または溶媒和物(本明細書においては、これらを総称して「製薬学的に許容される塩」という)で用いられ得る。適当な酸付加塩には、例えば、コハク酸、臭化水素酸、酢酸、フマル酸、マレイン酸、メタンスルホン酸、乳酸、リン酸、塩酸、硫酸、酒石酸、クエン酸およびヒベンズ酸から選択される酸の塩が挙げられる。好ましくは、CYP2D6の強い阻害薬として分類されているキニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブまたはそれらの製薬学的に許容される塩であり、より好ましくは、副作用が少なく臨床において容易に用いられ得るキニジン、パロキセチン、ブプロピオン、フルオキセチン、またはそれらの製薬学的に許容される塩であり、キニジンは硫酸塩水和物として、パロキセチンは塩酸塩水和物として、ブプロピオンは塩酸塩として、フルオキセチンは塩酸塩として用いられることが好ましい。更により好ましくは、主薬効を示す有効用量と阻害作用を示す有効用量が10倍以上異なるキニジンまたはその製薬学的に許容される塩が挙げられる。 The “CYP2D6 inhibitor” in the present invention means a plasma exposure (AUC) of a CYP2D6 substrate drug susceptible to interaction in a clinical drug interaction test by 1.25 times or more, preferably 2 times or more, more preferably 5 times or more. It means a drug having an inhibitory ability to increase more than twice. As the “CYP2D6 inhibitor” in the present invention, specifically, “Drug Interaction Guideline for Drug Development and Proper Information Provision (Final Draft)” issued on July 8, 2014 or September 26, 2016 Drug Development and Drug Interactions: Table of of Substrates, Inhibitors and and Inducers Table 3-2: Examples of clinical inhibitors for P450-mediated metabolisms (for concomitant use priorical and D quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, which are classified as strong inhibitors of CYP2D6 in drug labeling), duloxetine, mirabegron, celecoxib, escitalopram, moclobemide, which are classified as moderate inhibitors. F Ruvoxamine, abiraterone, amiodarone, clobazam, cobicistat, desvenlafaxine, labetalol, lorcaserin, ritonavir, sertraline, vemurafenib, deramcyclane or pharmaceutically acceptable salts thereof, which are classified as weak inhibitors, are mentioned. As the “CYP2D6 inhibitor” in the present invention, any two or more of these drugs may be used in combination. Although these drugs have different main actions, they also have a CYP2D6 inhibitory action, and are therefore referred to as “CYP2D6 inhibitors” in the present specification. These are free bases or pharmaceutically acceptable acid addition salts and / or hydrates and / or solvates thereof (in the present specification, these are collectively referred to as “pharmaceutically acceptable salts”). Called). Suitable acid addition salts are selected, for example, from succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulfonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulfuric acid, tartaric acid, citric acid and hibenzic acid. Acid salts may be mentioned. Preferred are quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib or pharmaceutically acceptable salts thereof, which are classified as strong inhibitors of CYP2D6, and more preferably have few side effects and are easy to clinically Quinidine, paroxetine, bupropion, fluoxetine, or a pharmaceutically acceptable salt thereof, wherein quinidine is a sulfate hydrate, paroxetine is a hydrochloride hydrate, bupropion is a hydrochloride, and fluoxetine is a hydrate. It is preferably used as the hydrochloride salt. Even more preferably, quinidine or a pharmaceutically acceptable salt thereof, which is different from the effective dose exhibiting the main drug effect by 10 times or more the effective dose exhibiting the inhibitory effect, is exemplified.
 本発明における中枢性疾患の一つの態様としては、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害(過眠、特発性過眠障害、ナルコレプシーを含む)、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症が挙げられる。好ましくは、過眠、特発性過眠障害、またはナルコレプシーを含む睡眠障害が挙げられる。 As one embodiment of the central diseases in the present invention, Parkinson's disease, Parkinson's disease syndrome due to the use of antipsychotic agents, sleep disorders (including hypersomnia, idiopathic hypersomnia disorder, narcolepsy), chronic fatigue syndrome, neurodegeneration and central nervous system Fatigue associated with sexual disorders, overeating, addiction, or fibromyalgia. Preferred examples include hypersomnia, idiopathic hypersomnia disorder, or sleep disorders including narcolepsy.
 本発明における中枢性疾患の別の態様としては、精神障害の診断と統計マニュアルの第5版(Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition)(以下、DSM-5という。)による分類IVである抑うつ障害群(Depressive Disorders)、分類Vである不安症群(Anxiety Disorders)、または分類Iである神経発達症群(Neurodevelopmental Disorders)が挙げられる。 Another aspect of the central disease in the present invention is classification IV according to the 5th edition (Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition) of the diagnosis and statistical manual of mental disorders (hereinafter referred to as DSM-5). Depressive disorders (Depressive Disorders), category V anxiety (Anxiety Disorders), or category I neurodevelopmental disorders (Neurodevelopmental Disorders).
 抑うつ障害群(Depressive Disorders)としては、不安性の苦痛を伴う抑うつ障害群(Depressive Disorders With anxious distress)、混合性の特徴を伴う抑うつ障害群(Depressive Disorders With mixed features)、メランコリアの特徴を伴う抑うつ障害群(Depressive Disorders With melancholic features)、気分に一致する精神病性の特徴を伴う抑うつ障害群(Depressive Disorders With mood-congruent psychotic features)、気分に一致しない精神病性の特徴を伴う抑うつ障害群(Depressive Disorders With mood-incongruent psychotic features)、緊張病を伴う抑うつ障害群(Depressive Disorders With catatonia)、季節型抑うつ障害群(Depressive Disorders With seasonal pattern)、重篤気分調整症(Disruptive Mood Dysregulation Disorder)、うつ病(Major Depressive Disorder)、持続性抑うつ障害(気分変調症)(Persistent Depressive Disorder(Dysthymia))、月経前不快気分障害(Premenstrual Dysphoric Disorder)、または物質・医薬品誘発性抑うつ障害(Substance/Medication-Induced Depressive Disorder)が挙げられる。 Depressive Disorders include Depressive Disorders With With anxious distress, Depressive Disorders With Mixed features, and depression with melancholic features. Depressive Disorders With melancholicfeatures, Depressive Disorders With-moog-congruent psychoticfeatures, Depressive Disorders with non-mood psychotic features With-mood-incongruent psychotic features, Depressive Disorders With Catatonia, Depressive Disorders, With seasonal pattern, Severe Mood Dysregulation Disorder, Depression. Major Depressive Disorder), persistent depressive disorder (dysthymia) (Persiste nt Depressive Disorder (Dysthymia)), premenstrual dysphoric disorder (Premenstrual Dysphoric Disorder), or substance / drug-induced depressive disorder (Substance / Medication-Induced Depressive Disorder).
 不安症群(Anxiety Disorders)としては、分離不安症(Separation Anxiety Disorder)、限局性恐怖症(Specific Phobia)、社交不安症(社交恐怖)(Social Anxiety Disorder(Social Phobia))、パニック症(Panic Disorder)、または物質・医薬品誘発性不安症(Substance/Medication-Induced Anxiety Disorder)が挙げられる。 Anxiety Disorders include Separation Anxiety Disorder, Specific Phobia, Social Anxiety Disorder (Social Phobia), and Panic Disorder. ), Or substance / drug-induced anxiety disorder (Substance / Medication-Induced Anxiety Disorder).
 神経発達症群(Neurodevelopmental Disorders)としては、自閉症スペクトラム症(Autism Spectrum Disorder)、注意欠如・多動症(Attention-Deficit/Hyperactivity Disorder)、限局性学習症(Specific Learning Disorder)、知的能力障害(Intellectual Disability)、言語症(Language Disorder)、語音症(Speech Sound Disorder)、小児期発症流暢症(Childhood-Onset Fluency Disorder(Stuttering))、社会的(語用論的)コミュニケーション症(Social(Pragmatic)Communication Disorder)、発達性協調運動症(Developmental Coordination Disorder)、チック症群(Tic Disorders)が挙げられる。 Neurodevelopmental Disorders include Autism Spectrum Disorder, Attention-Deficit / Hyperactivity Disorder, Specific Learning Disorder, and intellectual disability IntellectualDisability), Language Disorder, Speech Sound Disorder, Childhood-Onset Fluency Disorder (Stuttering), Social (Pragmatic) Communication (Pragmatic) Communication Disorder), Developmental Coordination Disorder, Tic Disorders.
 本発明における中枢性疾患の別の態様としては、小児における抑うつ障害群、または小児における不安症群が挙げられる。
 小児における抑うつ障害群としては、小児における不安性の苦痛を伴う抑うつ障害群、小児における混合性の特徴を伴う抑うつ障害群、小児におけるメランコリアの特徴を伴う抑うつ障害群、小児における気分に一致する精神病性の特徴を伴う抑うつ障害群、小児における気分に一致しない精神病性の特徴を伴う抑うつ障害群、小児における緊張病を伴う抑うつ障害群、小児における季節型抑うつ障害群、小児における重篤気分調整症、小児におけるうつ病、小児における持続性抑うつ障害(気分変調症)、小児における月経前不快気分障害、または小児における物質・医薬品誘発性抑うつ障害が挙げられる。
 小児における不安症群としては、小児における分離不安症、小児における限局性恐怖症、小児における社交不安症(社交恐怖)、小児におけるパニック症、または小児における物質・医薬品誘発性不安症が挙げられる。
Another aspect of the central diseases in the present invention includes a depressive disorder group in children or anxiety group in children.
Depression disorders in children include depressive disorders with anxious distress in children, depressive disorders with mixed features in children, depressive disorders with melancholic features in children, and psychosis consistent with mood in children. Depressive disorder group with sexual characteristics, depressive disorder group with psychotic features that do not match mood in children, depressive disorder group with catatonic disorders in children, seasonal depressive disorder group in children, severe mood regulation in children , Depression in children, persistent depressive disorder in children (dysthymia), premenstrual dysphoric disorder in children, or substance-drug-induced depressive disorder in children.
Anxiety groups in children include isolated anxiety in children, localized phobia in children, social anxiety in children (social phobia), panic disorder in children, or substance-drug-induced anxiety in children.
 本発明で用いる「小児」とは、概ね6歳~15歳を意味する。本発明による、チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬とを併用することによる、抑うつ障害群、不安症群、または神経発達症群の治療剤もしくは予防剤は、即効性があり、持続性があり、依存性を形成せず、自殺企図が起こらない安全な医薬であるため、成人患者だけでなく小児患者にも適用可能である。 The term “children” used in the present invention generally means 6 to 15 years old. The therapeutic or prophylactic agent for a depressive disorder group, anxiety group, or neurodevelopmental group by combining tipepidine or a pharmaceutically acceptable salt thereof with a CYP2D6 inhibitor according to the present invention has immediate effect. It is a safe medicine that is effective, persistent, does not form addiction, and does not cause suicide attempts, and is therefore applicable not only to adult patients but also to pediatric patients.
 本発明における中枢性疾患の別の態様としては、DSM-5による分類VIである強迫症および関連症群(Obsessive-Compulsive and Related Disorders)、または分類VIIである心的外傷およびストレス因関連障害群(Trauma- and Stressor-Related Disorders)が挙げられる。 As another embodiment of the central disease in the present invention, a group VI according to DSM-5 is obsessive-compulsive and related disorders (Obsessive-Compulsive and Related Related Disorders), or a group VII of trauma and stress-related disorders. (Trauma- and Stressor-Related Disorders).
 強迫症および関連症群としては、強迫症(Obsessive-Compulsive Disorder)、醜形恐怖症(Body Dysmorphic Disorder)、ためこみ症(Hoarding Disorder)、抜毛症(Trichotillomania(Hair-Pulling Disorder))、または皮膚むしり症(Excoriation(Skin-Picking)Disorder)が挙げられる。 Obsessive-compulsive Disorder, Obsessive-Compulsive Disorder, Body Dysmorphic Disorder, Hoarding Disorder, Trichotillomania (Hair-Pulling Disorder), or skin Exclusion (Skin-Picking) Disorder is mentioned.
 心的外傷およびストレス因関連障害群としては、心的外傷後ストレス障害(6 歳以下の子どもの心的外傷後ストレス障害を含む)(Posttraumatic Stress Disorder)、反応性アタッチメント障害(Reactive Attachment Disorder)、脱抑制型対人交流障害(Disinhibited Social Engagement Disorder)、急性ストレス障害(Acute Stress Disorder)、または適応障害(Adjustment Disorders)が挙げられる。 Post-traumatic stress disorder (including post-traumatic stress disorder in children under 6 years old) (Posttraumatic StressDisorder), reactive attachment disorder (Reactive AttachmentDisorder), Disinhibited social interaction disorder (Disinhibited Social Engagement Disorder), acute stress disorder (Acute Stress Disorder), or adjustment disorder (Adjustment Disorders).
 本発明で用いる「治療上の有効量」とは、組織、系、動物またはヒトにおいて、研究者または医師によって要求される生物学的または医薬的応答を誘発する薬物または医薬の量を意味する。
 本発明で用いる「治療」および「処置」とは、疾患のあらゆる治療(例えば、症状の改善、症状の軽減、症状の進行の抑制など)、並びに疾患のあらゆる予防(例えば、疾患の発症および/または進行の阻止)が含まれる。
As used herein, a "therapeutically effective amount" means an amount of a drug or drug that elicits a biological or pharmaceutical response required by a researcher or physician in a tissue, system, animal or human.
As used in the present invention, “treatment” and “treatment” refer to any treatment of a disease (eg, amelioration of symptoms, alleviation of symptoms, suppression of progression of symptoms, etc.) and any prevention of diseases (eg, onset and / or development of a disease). Or prevention of progress) is included.
 本発明で用いる「医薬組成物」、「医薬製剤」および「医薬製品」とは、特定の量で特定の成分を含む製品、並びに特定の量で特定の成分の組み合わせから直接的または間接的に生じるあらゆる製品を包含することを意図している。 As used herein, "pharmaceutical composition," "pharmaceutical formulation," and "pharmaceutical product" refer to products that contain the specified ingredients in the specified amounts, as well as directly or indirectly from combinations of the specified ingredients in the specified amounts. It is intended to encompass any product that results.
 本発明で用いる睡眠障害の定義とは、日本睡眠学会に従えば、「日中に過剰な眠気または実際に眠り込むことが毎日の様に繰り返して見られる状態で、少なくとも1ケ月間は持続し、そのため社会生活または職業的機能が妨げられ、あるいは自らが苦痛であると感じること。ただし一回の持続期間が1ヵ月より短くても繰り返して過眠期がみられるものも含む」である。 According to the Japanese Society of Sleep, the definition of sleep disorder used in the present invention is "a state in which excessive sleepiness or actual falling into sleep during the day is seen repeatedly every day, which lasts for at least one month, As a result, social life or occupational function is disturbed, or one feels distressed, even if the duration of one session is shorter than one month and repeated sleep periods are observed. ”
 本発明のチペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬との組み合わせにおいて、両剤は別々に投与してもよく、また1つの医薬組成物として一緒に投与してもよい。また、本発明の組み合わせの一方の成分を他方の成分に対して先に、同時に、または後に投与してもよい。これらの成分は、単一製剤形または分離した製剤形での医薬製剤に調製してもよく、キットとしてもよい。 In the combination of tipepidine or a pharmaceutically acceptable salt thereof of the present invention and a CYP2D6 inhibitor, both agents may be administered separately or may be administered together as one pharmaceutical composition. Also, one component of the combination of the invention may be administered prior to, simultaneously with, or after the other component. These components may be prepared as a pharmaceutical preparation in a single preparation form or separate preparation forms, or may be a kit.
 本発明の活性成分(チペピジンまたはその製薬学的に許容される塩および/またはCYP2D6阻害薬)は、経口、非経口(例えば、筋肉内、腹腔内、静脈内、経皮または皮下注射またはインプラント)、バッカル、経鼻、膣、直腸、舌下または局所(例えば、点眼)的な経路で投与してもよく、各投与経路に適した通常の無毒性の医薬的に許容される担体、補助剤、ベヒクルを含んだ適当な製剤中に、単独であるいは他の併用可能な薬剤と一緒に製剤化されていてもよい。 The active ingredient of the present invention (tipepidine or a pharmaceutically acceptable salt thereof and / or a CYP2D6 inhibitor) is orally or parenterally (for example, intramuscular, intraperitoneal, intravenous, transdermal or subcutaneous injection or implant). , Buccal, nasal, vaginal, rectal, sublingual or topical (eg, eye drop) route, and any conventional non-toxic pharmaceutically acceptable carrier or adjuvant suitable for each route of administration Alternatively, it may be formulated alone or in combination with other drugs that can be used in combination in a suitable formulation containing a vehicle.
 本発明の経口投与に適した医薬製品は、本発明の活性化合物であるチペピジンその製薬学的に許容される塩および/またはCYP2D6阻害薬を、活性成分の個々の必要性に適応した投与量で普通の投与形態、例えば錠剤、カプセル錠、シロップ剤、懸濁液等の型で経口的に投与することができる。あるいは、その溶液、乳剤、懸濁液等の液剤の型にしたものを注射の型で非経口的に投与することができ、またはパッチ剤にすることもできる。
 また、前記の適当な投与剤形は、許容される通常の担体、賦形剤、結合剤、安定化剤などに活性化合物を配合することにより製造することができる。また注射剤形で用いる場合には許容される緩衝剤、溶解補助剤、等張剤、pH調整剤等を添加することもできる。
Pharmaceutical products suitable for oral administration according to the invention comprise the active compound tipepidine of the invention, a pharmaceutically acceptable salt thereof and / or a CYP2D6 inhibitor, in a dose adapted to the individual needs of the active ingredient. It can be administered orally in the usual dosage forms, such as tablets, capsules, syrups, suspensions and the like. Alternatively, the solution, emulsion, suspension or the like in the form of a liquid preparation can be administered parenterally in the form of injection, or can be made into a patch.
Also, the above suitable dosage forms can be prepared by incorporating the active compound into an acceptable conventional carrier, excipient, binder, stabilizer and the like. When used in an injection dosage form, an acceptable buffering agent, solubilizing agent, isotonic agent, pH adjusting agent and the like can be added.
 本発明の活性成分の用量は特に制限されないが、治療薬の投与量、投与回数は、投与形態あるいは治療を要する疾病の病状の程度によって異なる。例えば、チペピジンまたはその製薬学的に許容される塩を単剤で投与する場合は、成人1日当り2-2000mg、好ましくは20-200mg、1回または数回に分けて経口または非経口で投与することができる。例えば、チペピジンまたはその製薬学的に許容される塩をCYP2D6阻害薬と併用して投与する場合は、成人1日当り2-2000mg、好ましくは20-200mg、1回または数回に分けて経口または非経口で投与することができ、併用するCYP2D6阻害薬としては、成人1日当り1-1000mg、好ましくは1-300mg、1回または数回に分けて経口または非経口で投与する事ができる。CYP2D6阻害薬として、例えばキニジンまたはその製薬学的に許容される塩は、成人1日当り1-200mg、好ましくは1-50mg、1回または数回に分けて経口または非経口で投与する事ができる。CYP2D6阻害薬として、例えばパロキセチンまたはその製薬学的に許容される塩は、成人1日当り1-200mg、好ましくは1-50mg、1回または数回に分けて経口または非経口で投与する事ができる。CYP2D6阻害薬として、例えばブプロピオンまたはその製薬学的に許容される塩は、成人1日当り1-1000mg、好ましくは1-300mg、1回または数回に分けて経口または非経口で投与する事ができる。CYP2D6阻害薬として、例えばフルオキセチンまたはその製薬学的に許容される塩は、成人1日当り1-200mg、好ましくは1-50mg、1回または数回に分けて経口または非経口で投与する事ができる。 The dose of the active ingredient of the present invention is not particularly limited, but the dose and frequency of administration of the therapeutic agent vary depending on the administration form or the degree of disease state requiring treatment. For example, when tipepidine or a pharmaceutically acceptable salt thereof is administered as a single agent, 2-2000 mg, preferably 20-200 mg per day for an adult is orally or parenterally administered once or in several divided doses. be able to. For example, when tipepidine or a pharmaceutically acceptable salt thereof is administered in combination with a CYP2D6 inhibitor, the dose is 2-2000 mg, preferably 20-200 mg per day for an adult, orally or The CYP2D6 inhibitor may be orally administered, and the combined CYP2D6 inhibitor may be 1-1000 mg, preferably 1-300 mg per day for an adult, and may be orally or parenterally administered once or in several divided doses. As a CYP2D6 inhibitor, for example, quinidine or a pharmaceutically acceptable salt thereof can be administered orally or parenterally per day for an adult at 1 to 200 mg, preferably 1 to 50 mg, once or in several divided doses. .. As a CYP2D6 inhibitor, for example, paroxetine or a pharmaceutically acceptable salt thereof can be orally or parenterally administered to an adult per day at 1 to 200 mg, preferably 1 to 50 mg, once or in several divided doses. .. As a CYP2D6 inhibitor, for example, bupropion or a pharmaceutically acceptable salt thereof can be administered orally or parenterally in an amount of 1 to 1000 mg, preferably 1 to 300 mg per day for an adult, or once or in several divided doses. .. As a CYP2D6 inhibitor, for example, fluoxetine or a pharmaceutically acceptable salt thereof can be administered orally or parenterally in an adult daily dose of 1-200 mg, preferably 1-50 mg, once or in several divided doses. ..
 チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬を併用して投与する際のチペピジン(フリー体換算):CYP2D6阻害薬(フリー体換算)の重量比として、1:500~2000:1が挙げられる。なお、「フリー体換算」とは、主薬をフリー体の形態で用いた場合はそのままの重量、主薬を製薬学的に許容される塩の形態で用いた場合は、塩の重量を除いたフリー体の重量として換算した重量をいう。
 例えば、CYP2D6阻害薬がキニジンまたはその製薬学的に許容される塩である場合、チペピジンまたはその製薬学的に許容される塩とキニジンまたはその製薬学的に許容される塩を併用して投与する際のチペピジン(フリー体換算):キニジン(フリー体換算)の重量比として、1:100~2000:1が挙げられる。例えば、CYP2D6阻害薬がパロキセチンまたはその製薬学的に許容される塩である場合、チペピジンまたはその製薬学的に許容される塩とパロキセチンまたはその製薬学的に許容される塩を併用して投与する際のチペピジン(フリー体換算):パロキセチンの重量比として、1:100~2000:1が挙げられる。例えば、CYP2D6阻害薬がブプロピオンまたはその製薬学的に許容される塩である場合、チペピジンまたはその製薬学的に許容される塩とブプロピオンまたはその製薬学的に許容される塩を併用して投与する際のチペピジン(フリー体換算):ブプロピオンの重量比として、1:500~2000:1が挙げられる。例えば、CYP2D6阻害薬がフルオキセチンまたはその製薬学的に許容される塩である場合、チペピジンまたはその製薬学的に許容される塩とフルオキセチンまたはその製薬学的に許容される塩を併用して投与する際のチペピジン(フリー体換算):フルオキセチンの重量比として、1:100~2000:1が挙げられる。
The weight ratio of tipepidine (free form equivalent): CYP2D6 inhibitor (free form equivalent) when administering in combination with tipepidine or a pharmaceutically acceptable salt thereof and the CYP2D6 inhibitor is 1: 500 to 2000: 1. Is mentioned. The term "free form equivalent" means the weight as it is when the main drug is used in the free form, and the free weight excluding the weight of the salt when the main drug is used in the form of a pharmaceutically acceptable salt. The weight converted as the weight of the body.
For example, when the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof, tipepidine or a pharmaceutically acceptable salt thereof and quinidine or a pharmaceutically acceptable salt thereof are administered in combination. In this case, the weight ratio of tipepidine (free body conversion): quinidine (free body conversion) is 1: 100 to 2000: 1. For example, when the CYP2D6 inhibitor is paroxetine or a pharmaceutically acceptable salt thereof, tipepidine or a pharmaceutically acceptable salt thereof and paroxetine or a pharmaceutically acceptable salt thereof are administered in combination. In this case, the weight ratio of tipepidine (calculated as free form): paroxetine is 1: 100 to 2000: 1. For example, when the CYP2D6 inhibitor is bupropion or a pharmaceutically acceptable salt thereof, tipepidine or a pharmaceutically acceptable salt thereof and bupropion or a pharmaceutically acceptable salt thereof are administered in combination. In this case, the weight ratio of tipepidine (free form conversion): bupropion is 1: 500 to 2000: 1. For example, when the CYP2D6 inhibitor is fluoxetine or a pharmaceutically acceptable salt thereof, tipepidine or a pharmaceutically acceptable salt thereof and fluoxetine or a pharmaceutically acceptable salt thereof are administered in combination. In this case, the weight ratio of tipepidine (free form conversion): fluoxetine is 1: 100 to 2000: 1.
 別の態様として、チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬を併用して投与する際のチペピジン(フリー体換算)の成人1日当りの下限値として、2~20mgの範囲の任意の重量、例えば、2mg、4mg、8mg、16mg、または20mgが挙げられ、CYP2D6阻害薬の成人1日当りの下限値として、1~20mgの範囲の任意の重量、例えば、1mg、2mg、5mg、10mg、または20mgが挙げられる。チペピジン(フリー体換算):CYP2D6阻害薬の重量比の下限値としては、例えば1:500、1:250、1:100、1:50、1:25、1:10、1:5、1:2が挙げられる。
 例えば、CYP2D6阻害薬がキニジンまたはその製薬学的に許容される塩である場合、キニジン(フリー体換算)の成人1日当りの下限値として、1mg、2mg、または5mgが挙げられる。チペピジン(フリー体換算):キニジン(フリー体換算)の重量比の下限値としては、1:100、1:50、1:25、1:10、1:5または1:2が挙げられる。
 例えば、CYP2D6阻害薬がパロキセチンまたはその製薬学的に許容される塩である場合、パロキセチン(フリー体換算)の成人1日当りの下限値として、1mg、2mg、または5mgが挙げられる。チペピジン(フリー体換算):パロキセチン(フリー体換算)の重量比の下限値としては、1:100、1:50、1:25、1:10、1:5または1:2が挙げられる。
 例えば、CYP2D6阻害薬がブプロピオンまたはその製薬学的に許容される塩である場合、ブプロピオン(フリー体換算)の成人1日当りの下限値として、1mg、10mg、または20mgが挙げられる。チペピジン(フリー体換算):ブプロピオン(フリー体換算)の重量比の下限値としては、1:500、1:300、1:150、1:100、1:50または1:25が挙げられる。
 例えば、CYP2D6阻害薬がフルオキセチンまたはその製薬学的に許容される塩である場合、フルオキセチン(フリー体換算)の成人1日当りの下限値として、1mg、2mg、または5mgが挙げられる。チペピジン(フリー体換算):フルオキセチン(フリー体換算)の重量比の下限値としては、1:100、1:50、1:25、1:10、1:5または1:2が挙げられる。
In another embodiment, the lower limit value of tipepidine (free body conversion) per day for adults when tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor are administered in combination is in the range of 2 to 20 mg. , For example, 2 mg, 4 mg, 8 mg, 16 mg, or 20 mg, and the lower limit of the daily dose of the CYP2D6 inhibitor for an adult is any weight in the range of 1 to 20 mg, for example, 1 mg, 2 mg, 5 mg, 10 mg. , Or 20 mg. The lower limit of the weight ratio of tipepidine (free form conversion): CYP2D6 inhibitor is, for example, 1: 500, 1: 250, 1: 100, 1:50, 1:25, 1:10, 1: 5, 1: 2.
For example, when the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof, 1 mg, 2 mg, or 5 mg can be mentioned as the lower limit of quinidine (free form conversion) per day for an adult. The lower limit of the weight ratio of tipepidine (converted free form): quinidine (converted free form) may be 1: 100, 1:50, 1:25, 1:10, 1: 5 or 1: 2.
For example, when the CYP2D6 inhibitor is paroxetine or a pharmaceutically acceptable salt thereof, the lower limit of paroxetine (free body conversion) for an adult per day is 1 mg, 2 mg, or 5 mg. The lower limit of the weight ratio of tipepidine (converted to free form): paroxetine (converted to free form) includes 1: 100, 1:50, 1:25, 1:10, 1: 5 or 1: 2.
For example, when the CYP2D6 inhibitor is bupropion or a pharmaceutically acceptable salt thereof, 1 mg, 10 mg, or 20 mg may be mentioned as the lower limit of bupropion (free form conversion) per day for an adult. The lower limit of the weight ratio of tipepidine (converted free form): bupropion (converted free form) includes 1: 500, 1: 300, 1: 150, 1: 100, 1:50 or 1:25.
For example, when the CYP2D6 inhibitor is fluoxetine or a pharmaceutically acceptable salt thereof, the lower limit of fluoxetine (free form conversion) per day for an adult is 1 mg, 2 mg, or 5 mg. The lower limit of the weight ratio of tipepidine (free form conversion): fluoxetine (free form conversion) may be 1: 100, 1:50, 1:25, 1:10, 1: 5 or 1: 2.
 別の態様として、チペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬を併用して投与する際のチペピジン(フリー体換算)の成人1日当りの上限値として、200~2000mgの範囲の任意の重量、例えば、200mg、400mg、800mg、1600mg、または2000mgが挙げられ、CYP2D6阻害薬の成人1日当りの上限値として、50~1000mgの範囲の任意の重量、例えば、50mg、100mg、200mg、500mg、または1000mgが挙げられる。チペピジン(フリー体換算):CYP2D6阻害薬の重量比の上限値としては、例えば2000:1、1000:1、800:1、400:1、200:1、100:1、50:1、25:1または10:1が挙げられる。
 例えば、CYP2D6阻害薬がキニジンまたはその製薬学的に許容される塩である場合、キニジン(フリー体換算)の成人1日当りの上限値として、50mg、100mg、または200mgが挙げられる。チペピジン(フリー体換算):キニジン(フリー体換算)の重量比の上限値としては、2000:1、1000:1、400:1、200:1、100:1、50:1、25:1または10:1が挙げられる。
 例えば、CYP2D6阻害薬がパロキセチンまたはその製薬学的に許容される塩である場合、パロキセチン(フリー体換算)の成人1日当りの上限値として、50mg、100mg、または200mgが挙げられる。チペピジン(フリー体換算):パロキセチン(フリー体換算)の重量比の上限値としては、2000:1、1000:1、400:1、200:1、100:1、50:1、25:1または10:1が挙げられる。
 例えば、CYP2D6阻害薬がブプロピオンまたはその製薬学的に許容される塩である場合、ブプロピオン(フリー体換算)の成人1日当りの上限値として、300mg、600mg、または1000mgが挙げられる。チペピジン(フリー体換算):ブプロピオン(フリー体換算)の重量比の上限値としては、2000:1、1000:1、400:1、200:1、100:1、50:1、25:1または10:1が挙げられる。
 例えば、CYP2D6阻害薬がフルオキセチンまたはその製薬学的に許容される塩である場合、フルオキセチン(フリー体換算)の成人1日当りの上限値として、50mg、100mg、または200mgが挙げられる。チペピジン(フリー体換算):フルオキセチン(フリー体換算)の重量比の上限値としては、2000:1、1000:1、400:1、200:1、100:1、50:1、25:1または10:1が挙げられる。
In another embodiment, the upper limit of the amount of tipepidine (free body conversion) per day for an adult when the tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor are administered in combination, is in the range of 200 to 2000 mg. , For example, 200 mg, 400 mg, 800 mg, 1600 mg, or 2000 mg, and the upper limit of the daily dose of the CYP2D6 inhibitor for an adult is any weight in the range of 50 to 1000 mg, for example, 50 mg, 100 mg, 200 mg, 500 mg. , Or 1000 mg. The upper limit of the weight ratio of tipepidine (free form conversion): CYP2D6 inhibitor is, for example, 2000: 1, 1000: 1, 800: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1.
For example, when the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof, 50 mg, 100 mg, or 200 mg may be mentioned as the upper limit of quinidine (free body conversion) per day for an adult. The upper limit of the weight ratio of tipepidine (converted free form): quinidine (converted free form) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
For example, when the CYP2D6 inhibitor is paroxetine or a pharmaceutically acceptable salt thereof, 50 mg, 100 mg, or 200 mg may be mentioned as the upper limit of paroxetine (free form conversion) per day for an adult. The upper limit of the weight ratio of tipepidine (converted free form): paroxetine (converted free form) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
For example, when the CYP2D6 inhibitor is bupropion or a pharmaceutically acceptable salt thereof, the upper limit of bupropion (free body conversion) for an adult per day is 300 mg, 600 mg, or 1000 mg. The upper limit of the weight ratio of tipepidine (free form conversion): bupropion (free form conversion) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
For example, when the CYP2D6 inhibitor is fluoxetine or a pharmaceutically acceptable salt thereof, 50 mg, 100 mg, or 200 mg may be mentioned as the upper limit of fluoxetine (free body conversion) per day for an adult. The upper limit of the weight ratio of tipepidine (free form conversion): fluoxetine (free form conversion) is 2000: 1, 1000: 1, 400: 1, 200: 1, 100: 1, 50: 1, 25: 1 or 10: 1 can be mentioned.
 本発明の医薬組成物は、経口投与のための内服用固形剤、内服用液剤および、非経口投与のための注射剤、外用剤、坐剤、吸入剤、経鼻剤等として用いられる。経口投与のための内服用固形剤には、錠剤、丸剤、カプセル剤、散剤、顆粒剤等が含まれる。カプセル剤には、ハードカプセルおよびソフトカプセルが含まれる。また錠剤には舌下錠、口腔内貼付錠、口腔内速崩壊錠などが含まれる。 The pharmaceutical composition of the present invention is used as a solid preparation for oral administration, a liquid preparation for oral administration, and an injection, an external preparation, a suppository, an inhalant, a nasal preparation, etc. for parenteral administration. The solid preparations for oral administration include tablets, pills, capsules, powders and granules. Capsules include hard capsules and soft capsules. In addition, tablets include sublingual tablets, buccal tablets, and rapidly disintegrating buccal tablets.
 このような内服用固形剤においては、ひとつまたはそれ以上の活性物質はそのままか、または賦形剤(ラクトース、マンニトール、グルコース、結晶セルロース、デキストリン、デンプン等)、結合剤(ヒドロキシプロピルセルロース、ポリビニルピロリドン、メタケイ酸アルミン酸マグネシウム等)、崩壊剤(繊維素グリコール酸カルシウム等)、滑沢剤(ステアリン酸マグネシウム、タルク等)、安定化剤、溶解補助剤(グルタミン酸、アスパラギン酸等)等と混合され、常法に従って製剤化して用いられる。また、必要によりコーティング剤(白糖、ゼラチン、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロースフタレート等)で被覆していてもよいし、また2以上の層で被覆していてもよい。さらにゼラチンのような吸収されうる物質のカプセルも包含される。また、必要に応じて常用される防腐剤、抗酸化剤、着色剤、甘味剤等の添加物を加えることもできる。 In such solid oral preparations, one or more active substances may be used as they are, or excipients (lactose, mannitol, glucose, crystalline cellulose, dextrin, starch, etc.), binders (hydroxypropylcellulose, polyvinylpyrrolidone). , Magnesium metasilicate, etc.), disintegrant (calcium fibrin glycolate, etc.), lubricant (magnesium stearate, talc, etc.), stabilizer, solubilizer (glutamic acid, aspartic acid, etc.), etc. It is used by formulating it according to a conventional method. If necessary, it may be coated with a coating agent (sucrose, gelatin, hydroxypropylcellulose, hydroxypropylmethylcellulose phthalate, etc.), or may be coated with two or more layers. Also included are capsules of absorbable material such as gelatin. In addition, additives such as preservatives, antioxidants, colorants, sweeteners and the like which are commonly used can be added if necessary.
 舌下錠、口腔内貼付錠、口腔内速崩壊錠、経口投与のための内服用液剤は、各剤形に適した通常の担体、賦形剤、結合剤、安定化剤などと混合して、公知の方法に準じて製造される。 Sublingual tablets, buccal patches, rapidly disintegrating buccal tablets, and oral liquid preparations for oral administration are mixed with usual carriers, excipients, binders, stabilizers, etc. suitable for each dosage form. It is manufactured according to a known method.
 非経口投与のための外用剤の剤形には、例えば、軟膏剤、ゲル剤、クリーム剤、湿布剤、貼付剤、リニメント剤、噴霧剤、吸入剤、スプレー剤、エアゾル剤、点眼剤、および点鼻剤等が含まれる。これらはひとつまたはそれ以上の活性物質を含み、公知の方法または通常使用されている処方により製造される。 The dosage form of the external preparation for parenteral administration includes, for example, ointments, gels, creams, poultices, patches, liniments, sprays, inhalants, sprays, aerosols, eye drops, and Includes nasal drops and the like. These contain one or more active substances and are produced by a known method or a commonly used formulation.
 湿布剤、貼付剤、非経口投与のための注射剤は、各剤形に適した通常の医薬添加剤と混合して、公知または通常使用されている処方により製造される。 A poultice, a patch, and an injection for parenteral administration are manufactured by a known or commonly used formulation by mixing with a usual pharmaceutical additive suitable for each dosage form.
 本明細書において、「有効量」とは、中枢性疾患の進行を完全または部分的に阻害するか、あるいは、中枢性疾患の1以上の症状を少なくとも部分的に緩和する、チペピジンまたはその製薬学的に許容される塩の量、またはチペピジンまたはその製薬学的に許容される塩とCYP2D6阻害薬との組合せの量である。有効量は、治療的または予防的に有効な量であり得る。有効量は、患者の年齢および性別、処置される状態、状態の重度、ならびに求められている結果などにより決定される。所定の患者について、有効量は、当業者に知られる方法によって決定することができる。 As used herein, the term “effective amount” refers to tipepidine or a pharmaceutical preparation thereof, which completely or partially inhibits the progression of central diseases, or at least partially alleviates one or more symptoms of central diseases. Or a combination of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor. An effective amount can be a therapeutically or prophylactically effective amount. An effective amount will be determined by the age and sex of the patient, the condition being treated, the severity of the condition, the results sought, and the like. For a given patient, the effective amount can be determined by methods known to those of ordinary skill in the art.
実施例1:
ヒト肝細胞を用いたチペピジンのCYP代謝寄与の推定
 チペピジンのヒト肝細胞による代謝における、CYP代謝の寄与を以下の方法で確認した。ヒト凍結肝細胞はSekisui XenoTech社製を使用した。ヒト凍結肝細胞をKHB buffer中に100万cells/mLの濃度で播種し、そこにチペピジンを1μmol/Lの濃度で加えた。そこにCYP阻害薬である1-アミノベンゾトリアゾール(1-ABT)(2.5mmol/L)を加えた処置群と、加えない非処置群を設け、それぞれ37℃、5% COの条件下でインキュベーションし、1時間または2時間反応させた。反応後のチペピジンの残存率LC-MSで測定した。
Example 1:
Estimation of CYP metabolism contribution of tipepidine using human hepatocytes The contribution of CYP metabolism to the metabolism of tipepidine by human hepatocytes was confirmed by the following method. Human frozen hepatocytes used were those manufactured by Sekisui XenoTech. Human frozen hepatocytes were seeded in KHB buffer at a concentration of 1 million cells / mL, and tipepidine was added thereto at a concentration of 1 μmol / L. A treatment group to which 1-aminobenzotriazole (1-ABT) (2.5 mmol / L), which is a CYP inhibitor, was added and a non-treatment group to which no CYP inhibitor was added were provided, and the conditions were 37 ° C. and 5% CO 2 And incubated for 1 hour or 2 hours. The residual rate of tipepidine after the reaction was measured by LC-MS.
 結果を図1に示した。ヒト肝細胞によるチペピジンの代謝はCYP阻害薬である1-ABTにより阻害されることが示され、チペピジンのヒト肝臓中での代謝は主にCYPが担うことが明らかとなった。 The results are shown in Figure 1. It was shown that the metabolism of tipepidine by human hepatocytes was inhibited by 1-ABT, which is a CYP inhibitor, and it was revealed that CYP is mainly responsible for the metabolism of tipepidine in human liver.
実施例2:
ヒト肝ミクロソームを用いたチペピジンの各種CYP代謝寄与率の算出
 ヒト肝ミクロソームを用いてチペピジンの代謝に関与する各種CYPの代謝寄与率を算出した。ヒト肝ミクロソームはSekisui XenoTech社製を使用した。50mmol/Lのリン酸緩衝液(pH7.4)中にヒト肝ミクロソームを0.1mg/mL、NADPHを3mmol/L、チペピジンを0.1μmol/Lの濃度になるように加え、さらに各種CYPの選択的阻害薬を以下に示す濃度になるように混合し、37℃で30分間インキュベーションした。
Example 2:
Calculation of Various CYP Metabolism Contribution Ratios of Tipepidine Using Human Liver Microsomes The metabolic contribution ratios of various CYPs involved in the metabolism of tipepidine were calculated using human liver microsomes. Human liver microsomes used were manufactured by Sekisui XenoTech. Human liver microsomes were added at a concentration of 0.1 mg / mL, NADPH at 3 mmol / L, and tipepidine at a concentration of 0.1 μmol / L in a 50 mmol / L phosphate buffer (pH 7.4), and various CYPs were added. The selective inhibitors were mixed at the concentrations shown below and incubated at 37 ° C for 30 minutes.
各種CYPの選択的阻害薬濃度
フラフィリン(furafylline;CYP1A2):10μmol/L、モンテルカスト(montelukast;CYP2C8):10μmol/L、スルファフェナゾール(sulfaphenazole;CYP2C9):10μmol/L、N-ベンジルニルバノール(N-benzylnirvanol;CYP2C19):5μmol/L、キニジン(quinidine;CYP2D6):1μmol/L、アザムリン(azamulin;CYP3A4):5μmol/L
Concentration of selective inhibitor of various CYPs Furafylline (CYP1A2): 10 μmol / L, montelukast (CYP2C8): 10 μmol / L, sulfaphenazole (CYP2C9): 10 μmol / L, N-benzylnilvanol ( N-benzylnirvanol; CYP2C19): 5 μmol / L, quinidine (CYP2D6): 1 μmol / L, azamulin (CYP3A4): 5 μmol / L
 反応後のサンプル中のチペピジンの残存率をLC-MSにて測定し、以下の式から代謝クリアランス、各種CYPの代謝寄与率を算出した。
・代謝クリアランス(μL/min/mg protein)=-LN(残存率)/30/0.1×1000
・代謝寄与率(%)=(1-各種CYP阻害薬存在時の代謝クリアランス/阻害薬非存在時の代謝クリアランス)×100
The residual rate of tipepidine in the sample after the reaction was measured by LC-MS, and the metabolic clearance and the metabolic contribution rate of various CYPs were calculated from the following formulas.
・ Metabolic clearance (μL / min / mg protein) =-LN (residual rate) /30/0.1×1000
Metabolic contribution rate (%) = (1-metabolic clearance in the presence of various CYP inhibitors / metabolic clearance in the absence of inhibitors) x 100
 結果を表1に示した。表1はヒト肝ミクロソーム中チペピジン代謝における各種CYP阻害時の残存率と代謝クリアランス及び代謝クリアランスから計算した各種CYPの代謝寄与率を示したものである。チペピジンは主にCYP2D6により代謝されることが初めて明らかとなり、その寄与率は83%と算出された。
Figure JPOXMLDOC01-appb-T000001
The results are shown in Table 1. Table 1 shows the survival rate at the time of inhibition of various CYPs in the metabolism of tipepidine in human liver microsomes, the metabolic clearance, and the metabolic contribution rate of various CYPs calculated from the metabolic clearance. For the first time, it was revealed that tipepidine was mainly metabolized by CYP2D6, and its contribution rate was calculated to be 83%.
Figure JPOXMLDOC01-appb-T000001
実施例3:
チペピジン単独、CYP2D6阻害薬とチペピジン併用時のヒトにおけるチペピジン血漿中濃度推移及びPKパラメータのシミュレーション
 チペピジンを単独で経口投与した際の血漿中濃度推移及びPKパラメータを、チペピジンを有効成分として含有する市販のアスベリン錠のインタビューフォーム(IF)に記載されている情報を以下の1-コンパートメントモデル式に当てはめて計算することで算出した。モデル式中の略称は血漿中濃度(C)、生物学的利用率(F)、投与量(Dose)、分布容積(Vd)、吸収速度定数(Ka)、消失速度定数(Kel)、投与後時間(t)、最高血漿中濃度到達時間(tmax)、最高血漿中濃度(Cmax)、血漿中濃度時間曲線下面積(AUC)、全身クリアランス(CL)、半減期(t1/2)、肝血流量(Q)、血漿蛋白結合率(fu)、肝固有クリアランス(CLint)とした。計算の仮定としてチペピジンの体内からの消失は主に肝臓中での代謝によるものとし、また消化管吸収は良好であり、さらに血液中濃度/血漿中濃度比は1とした。また肝血流量は84L/hという値を使用し、全身クリアランスと肝固有クリアランスの関係式としてパラレルチューブモデル式を使用した。
Example 3:
Simulation of Tipepidine Plasma Concentration Changes in Humans and PK Parameters in Combination with Tipepidine Alone, CYP2D6 Inhibitor and Tipepidine Combined Commercially-Containing Tipepidine as an Active Ingredient in Plasma Concentration Changes and PK Parameters after Oral Administration of Tipepidine It was calculated by applying the information described in the Asverin Tablet Interview Form (IF) to the following 1-compartment model formula. Abbreviations in the model formula are plasma concentration (C), bioavailability (F), dose (Dose), volume of distribution (Vd), absorption rate constant (Ka), elimination rate constant (Kel), post-dose Time (t), time to reach maximum plasma concentration (t max ), maximum plasma concentration (C max ), area under plasma concentration time curve (AUC), systemic clearance (CL), half-life (t 1/2 ) , Hepatic blood flow (Q), plasma protein binding rate (fu), and liver specific clearance (CL int ). As a calculation assumption, the elimination of tipepidine from the body was mainly due to metabolism in the liver, the gastrointestinal absorption was good, and the blood concentration / plasma concentration ratio was 1. A value of 84 L / h was used for the hepatic blood flow, and a parallel tube model formula was used as a relational expression between the whole body clearance and the hepatic clearance.
モデル式
C=F×Dose/Vd×Ka/(Ka-Kel)×(e-Kel×t-e-Ka×t
tmax=1/(Ka-Kel)×ln(Ka/Kel)
Cmax=F×Dose/Vd×(Ka/Kel)(Kel/(Ka-Kel))
AUC=F×Dose/CL
t1/2=0.693/Kel
Kel=CL/Vd
F=1-CL/Q
CL=Q×(1-e(-fu×CLint/Q)
Model formula
C = F × Dose / Vd × Ka / (Ka-Kel) × (e -Kel × t -e -Ka × t)
t max = 1 / (Ka-Kel) × ln (Ka / Kel)
C max = F × Dose / Vd × (Ka / Kel) (Kel / (Ka-Kel))
AUC = F × Dose / CL
t 1/2 = 0.693 / Kel
Kel = CL / Vd
F = 1-CL / Q
CL = Q × (1−e (-fu × CLint / Q) )
 さらに同一のモデルにおいてチペピジンの肝固有クリアランスが83%低下した場合、つまりCYP2D6による代謝が全て阻害された場合の計算を実施することでCYP2D6阻害薬とチペピジンを併用して経口投与した際のチペピジンの血漿中濃度推移及びPKパラメータを算出した。 Furthermore, in the same model, when the liver-specific clearance of tipepidine was reduced by 83%, that is, when the metabolism by CYP2D6 was completely inhibited, calculation was performed to determine the amount of tipepidine when CYP2D6 inhibitor and tipepidine were orally administered in combination. Changes in plasma concentration and PK parameters were calculated.
 ヒトにチペピジン単独で経口投与、またはCYP2D6阻害薬とチペピジンとを併用して経口投与した時のチペピジンの血漿中濃度推移のシミュレーション結果を図2に、PKパラメータのシミュレーション結果を表2に示した。チペピジン単独投与時と比較してCYP2D6阻害薬とチペピジン併用時のチペピジンのCmaxは最大3.1倍、AUCは最大9.4倍、t1/2は最大4.0倍に増加するとシミュレーションされ、チペピジンをCYP2D6阻害薬と併用することでチペピジンの血漿中曝露の上昇及び半減期の増加が期待できることが示された。またCYP2D6の強い阻害薬として分類されているキニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブはCYP2D6を少なくとも80%以上阻害することから、CYP2D6を80%~100%阻害するとして同様にシミュレーションすると、これらの阻害薬との併用時にチペピジンのCmaxは2.4~3.1倍、AUCは4.3~9.4倍、t1/2は2.2~4.0倍に増加すると計算された。
Figure JPOXMLDOC01-appb-T000002
FIG. 2 shows the simulation results of changes in the plasma concentration of tipepidine when orally administered to humans alone with tipepidine alone, or when orally administered with the combination of CYP2D6 inhibitor and tipepidine, and Table 2 shows the simulation results of PK parameters. It was simulated that the C max of tipepidine when the CYP2D6 inhibitor and tipepidine were combined was increased by a maximum of 3.1 times, the AUC was increased by a maximum of 9.4 times, and the t 1/2 was increased by a maximum of 4.0 times as compared with the case of administration of tipepidine alone. It was shown that the combination of tipepidine with a CYP2D6 inhibitor can be expected to increase plasma exposure and half-life of tipepidine. Moreover, since quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, and dacomitinib, which are classified as strong inhibitors of CYP2D6, inhibit CYP2D6 by at least 80% or more, the same simulation is performed assuming that CYP2D6 is inhibited by 80% to 100%. , When combined with these inhibitors, the C max of tipepidine increases by 2.4 to 3.1 times, the AUC increases by 4.3 to 9.4 times, and the t 1/2 increases by 2.2 to 4.0 times. calculated.
Figure JPOXMLDOC01-appb-T000002
実施例4:
PXBマウスにおけるチペピジン単独、CYP2D6阻害薬キニジンとチペピジン併用時のチペピジン血中動態の評価
 実施例3のシミュレーションで示したCYP2D6阻害薬併用によるチペピジンの血漿中曝露の上昇及び半減期の増加効果を動物を用いて検証する目的で、ヒト肝細胞移植マウスであるPXBマウスを用いたチペピジンの経口投与試験を実施した。なお薬物代謝には一般的に種差があることが知られており、各種CYPの代謝寄与率も異なることから、野生型のマウスやラットを用いるのではなく、ヒトの肝細胞を移植したPXBマウスを用いる方がヒトにおけるCYP2D6阻害薬の併用効果を検証することに適していると考えられる。ヒト肝臓移植マウスである雄性PXBマウス(18-20週齢)に、チペピジンヒベンズ酸塩20mg/kgを単独で経口投与、あるいはCYP2D6阻害薬のキニジン100mg/kgを経口投与した2時間後にチペピジンヒベンズ酸塩20mg/kgを経口投与した。チペピジンヒベンズ酸塩投与後15分、30分、1時間、3時間、6時間および24時間で血液を採取した。採取した血液から血漿を得、LC-MSにて血漿中チペピジン濃度を測定し、この濃度推移をPhoenix WinNonlin(Certara社)を用いてノンコンパートメント解析し、PKパラメータとしてCmax、AUC、t1/2を算出した。
Example 4:
Evaluation of Tipepidine Blood Kinetics of Tipepidine Alone in PXB Mice and in Combination with CYP2D6 Inhibitor Quinidine and Tipepidine. For the purpose of verification using the above, an oral administration test of tipepidine was carried out using PXB mice, which are human hepatocyte transplanted mice. It is known that there are species differences in drug metabolism in general, and the contribution ratio of various CYPs to metabolism is also different. Therefore, PXB mice transplanted with human hepatocytes are used instead of using wild-type mice or rats. It is considered that the use of is more suitable for verifying the combined effect of CYP2D6 inhibitors in humans. 20 mg / kg of tipepidine hibenzate alone was orally administered to male PXB mice (18-20 weeks old), which are human liver transplantation mice, or 100 mg / kg of CYP2D6 inhibitor quinidine was orally administered, and 2 hours after that, tipepidine hibenzic acid was administered. 20 mg / kg of salt was orally administered. Blood was collected at 15 minutes, 30 minutes, 1 hour, 3 hours, 6 hours, and 24 hours after administration of tipepidine hibenzate. Plasma was obtained from the collected blood, the plasma tipepidine concentration was measured by LC-MS, and the concentration transition was subjected to non-compartmental analysis using Phoenix WinNonlin (Certara), and C max , AUC, t 1 / as PK parameters. 2 was calculated.
 ヒト肝細胞移植マウス(PXBマウス)にチペピジンヒベンズ酸塩20mg/kgを単独で経口投与、またはCYP2D6阻害薬キニジン100mg/kgとチペピジンヒベンズ酸塩20mg/kgとを併用して経口投与した時のチペピジンの血漿中濃度推移を図3に、PKパラメータを表3に示した。チペピジンをCYP2D6阻害薬キニジンと併用投与することで、チペピジンのCmaxが1.5倍、AUCが3.2倍、t1/2が3.0倍に増加することが明らかとなり、ヒトにおいても血漿中曝露の上昇および半減期の増加が期待されることが示された。なお本実施例で示された上昇率は実施例3においてシミュレーションされた上昇率よりも低い値であったが、これはPXBマウスの肝臓が完全にはヒト化しておらず、マウスの残存肝細胞による代謝がキジニンの影響を受けなかったためと推察される。
Figure JPOXMLDOC01-appb-T000003
Tipepidine hibenzate 20 mg / kg alone orally administered to a human hepatocyte transplanted mouse (PXB mouse) or tipepidine when CYP2D6 inhibitor quinidine 100 mg / kg and tipepidine hibenzate 20 mg / kg are orally administered together 3 shows the changes in the plasma concentration of A. and PK parameters in Table 3. By co-administering tipepidine with CYP2D6 inhibitor quinidine, it was clarified that the C max of tipepidine was increased by 1.5 times, AUC was increased by 3.2 times, and t 1/2 was increased by 3.0 times. It has been shown that increased plasma exposure and increased half-life are expected. The rate of increase shown in this Example was lower than the rate of increase simulated in Example 3, but this was due to the fact that the liver of PXB mouse was not completely humanized and the residual hepatocytes of the mouse were It is presumed that the metabolism of the drug was not affected by chidinin.
Figure JPOXMLDOC01-appb-T000003
実施例5:
SDラットにおけるチペピジン単独、CYP阻害薬1-ABTとチペピジン併用時のチペピジン血中動態の評価
 実施例4によりPXBマウスにおいてチペピジンをCYP2D6阻害薬キニジンと併用することでチペピジンの血漿中曝露の上昇および半減期の増加が確認されたが、PXBマウスは薬理評価には適していない。よって薬理評価に適したラットにおけるCYP阻害薬併用による曝露上昇効果を確認する目的で、SDラットを用いたチペピジンの経口投与試験を実施した。なお各種CYPの代謝寄与率にヒトとラットに種差があることを考慮してCYP2D6阻害薬ではなく、全てのCYPを阻害する1-ABTを用いた。SD雄性ラット(7週齢)に溶媒、又はCYP阻害薬1-ABT100mg/kgを経口投与した。溶媒、又は1-ABT投与2時間後に、チペピジンヒベンズ酸塩20mg/kgを経口投与した。投与後30分、1時間、2時間、4時間、6時間で血液を採取した。採取した血液から血漿を得、LC-MSにて血漿中チペピジン濃度を測定し、この濃度推移をPhoenix WinNonlin(Certara社)を用いてノンコンパートメント解析し、PKパラメータとしてCmax、AUC、t1/2を算出した。
Example 5:
Evaluation of Tipepidine Blood Kinetics of Tipepidine Alone and CYP Inhibitor 1-ABT in Combination with Tipepidine in SD Rats According to Example 4, by combining tipepidine with CYP2D6 inhibitor quinidine in PXB mice, increase and halve in plasma exposure of tipepidine Although an increase in the period was confirmed, PXB mice are not suitable for pharmacological evaluation. Therefore, an oral administration test of tipepidine using SD rats was carried out for the purpose of confirming the exposure-increasing effect of the combined use of CYP inhibitors in rats suitable for pharmacological evaluation. Considering that there is a species difference in the contribution ratio of various CYPs between humans and rats, 1-ABT that inhibits all CYPs was used instead of the CYP2D6 inhibitor. A solvent or CYP inhibitor 1-ABT 100 mg / kg was orally administered to SD male rats (7 weeks old). Two hours after the administration of the solvent or 1-ABT, 20 mg / kg of tipepidine hibenzate was orally administered. Blood was collected 30 minutes, 1 hour, 2 hours, 4 hours, and 6 hours after administration. Plasma was obtained from the collected blood, the plasma tipepidine concentration was measured by LC-MS, and the concentration transition was subjected to non-compartmental analysis using Phoenix WinNonlin (Certara), and C max , AUC, t 1 / as PK parameters. 2 was calculated.
 SDラットにチペピジンヒベンズ酸塩20mg/kgを単独で経口投与、またはCYP阻害薬1-ABT100mg/kgとチペピジンヒベンズ酸塩20mg/kgとを併用して経口投与した時のチペピジンの血漿中濃度推移を図4に、PKパラメータを表4に示した。チペピジンを1-ABTと併用投与することでCmaxが16倍、AUCが93倍、t1/2が12倍に増加することが明らかとなった。
Figure JPOXMLDOC01-appb-T000004
Changes in plasma concentrations of tipepidine when SD rats were orally administered with 20 mg / kg of tipepidine hibenzate alone, or with orally administered 100 mg / kg of CYP inhibitor 1-ABT and 20 mg / kg of tipepidine hibenzate. In FIG. 4, PK parameters are shown in Table 4. It was revealed that coadministration of tipepidine with 1-ABT increased C max 16-fold, AUC 93-fold, and t 1/2 12-fold.
Figure JPOXMLDOC01-appb-T000004
実施例6:
チペピジンの中枢性副作用の検討
 新規環境下における自発運動量の評価は、中枢に作用する薬物の副作用評価として一般的に用いられている評価系である。SD雄性ラット(7週齢)32匹に溶媒、又はチペピジンヒベンズ酸塩25mg/kg、または80mg/kgを腹腔内投与した。投与30分後から90分間、ラットの自発運動量をSUPERMEX装置(室町機械)により測定した。
Example 6:
Examination of central side effects of tipepidine The evaluation of locomotor activity in a new environment is an evaluation system generally used for evaluating side effects of drugs acting on the central nervous system. Thirty-two SD male rats (7 weeks old) were intraperitoneally administered with a solvent or tipepidine hibenzate 25 mg / kg or 80 mg / kg. From 30 minutes after the administration, the spontaneous locomotor activity of the rats was measured for 90 minutes by a SUPERMEX apparatus (Muromachi Kikai).
 結果を図5に示した。チペピジンヒベンズ酸塩25mg/kg又は80mg/kgを腹腔内投与することにより、中枢性の副作用と考えらえる自発運動量が有意に増加していた。チペピジンヒベンズ酸塩25mg/kgの予測血中薬物フリー濃度(6.4nM)は、ヒトの臨床において抗うつ作用が認められた予測血中薬物フリー濃度(0.37nM)を超える超高用量であり、超高用量ではチペピジンが中枢性の副作用を示すことが初めて明らかとなった。 The results are shown in Figure 5. Intraperitoneal administration of tipepidine hibenzate 25 mg / kg or 80 mg / kg significantly increased the amount of locomotor activity considered to be a central side effect. The predicted blood drug-free concentration (6.4 nM) of tipepidine hibenzate 25 mg / kg was an extremely high dose exceeding the predicted blood drug-free concentration (0.37 nM), which was observed to have antidepressant activity in human clinical studies. For the first time, it became clear that tipepidine had central side effects at very high doses.
実施例7:
Wistarラットに対する強制水泳試験を用いたチペピジンの抗うつ様作用
 強制水泳試験は、抗うつ薬のスクリーニングをはじめとして、ラットやマウスのうつ病様行動を評価する行動試験として広く用いられている。Wistar雄性ラット(7週齢)36匹を試験前日に水泳訓練室に投入して環境に慣らした。その後、水温25±1度の水道水を満たした水槽に動物を投入して、15分間強制水泳訓練試験を行った。なお、抗うつ薬イミプラミン10mg/kgを水泳訓練前に皮下投与を行った。水泳後、動物を水槽から取り出し、水滴をふき取りケージに戻した。試験当日、体重測定を行い、水泳訓練室に搬入して環境に慣らした。イミプラミン投与群においては、イミプラミンを試験開始の1時間前及び5時間前に皮下投与を行い、チペピジン投与群においては、チペピジンヒベンズ酸塩8mg/kgまたは80mg/kgを試験開始の30分前に腹腔内投与を行った。水温25±1度の水道水を満たした水槽に動物を投入して、5分間強制水泳訓練試験を行い、前面からビデオカメラで撮影した。無動時間の測定は動画をブラインドした後、目視にて測定を行った。
Example 7:
Antidepressant-like Action of Tipepidine Using Forced Swim Test on Wistar Rats The forced swim test is widely used as a behavioral test to evaluate depression-like behavior of rats and mice, including screening for antidepressant drugs. Thirty-six Wistar male rats (7 weeks old) were placed in the swimming training room on the day before the test to get accustomed to the environment. Then, the animal was put into a water tank filled with tap water at a water temperature of 25 ± 1 degree, and a forced swimming training test was conducted for 15 minutes. The antidepressant drug imipramine 10 mg / kg was subcutaneously administered before swimming training. After swimming, the animals were removed from the aquarium and water droplets were wiped off and returned to their cages. On the day of the test, the body weight was measured and brought into a swimming training room to get accustomed to the environment. In the imipramine administration group, imipramine was subcutaneously administered 1 hour and 5 hours before the start of the test. In the tipepidine administration group, tipepidine hibenzate 8 mg / kg or 80 mg / kg was intraperitoneally administered 30 minutes before the start of the test. Internal administration was performed. The animal was placed in a water tank filled with tap water at a water temperature of 25 ± 1 ° C., a forced swimming training test was conducted for 5 minutes, and a video camera was photographed from the front. The immobility time was measured visually after blinding the moving image.
 結果を図6に示した。チペピジンヒベンズ酸塩8mg/kgまたは80mg/kgを腹腔内投与により、有意な無動時間の短縮を見出し、強い抗うつ様作用をチペピジンが有していることが明らかとなった。なお、チペピジンヒベンズ酸塩8mg/kgの血中薬物フリー濃度は0.93nMであり、ヒトの臨床試験において抗うつ作用が認められた予測血中薬物フリー濃度が0.37nMであることから、ヒトの臨床で認められた血中濃度以上に、より強い抗うつ作用を有していることが明らかとなった。 The results are shown in Figure 6. By intraperitoneal administration of tipepidine hibenzate 8 mg / kg or 80 mg / kg, it was found that immobility time was significantly shortened, and it was revealed that tipepidine has a strong antidepressant action. The blood drug-free concentration of tipepidine hibenzate 8 mg / kg was 0.93 nM, and the predicted blood drug-free concentration for which antidepressant action was observed in human clinical studies was 0.37 nM. It was revealed that it has a stronger antidepressant action than the blood concentration observed in the clinical trial.
実施例8:
チペピジン、又はチペピジン及びCYP阻害薬併用による中枢性副作用の検討
 SD雄性ラット(7週齢)に、溶媒又はCYP阻害薬1-ABT 100mg/kgを強制経口投与した。溶媒を投与したラット群を溶媒投与群、1-ABTを投与したラット群を1-ABT投与群とし、それぞれの群の溶媒又は1-ABT投与2時間後に、溶媒投与群においては溶媒又はチペピジンヒベンズ酸塩20mg/kgを投与し、1-ABT投与群においてはチペピジンヒベンズ酸塩20mg/kgを強制経口投与した。溶媒又はチペピジンヒベンズ酸塩20mg/kg投与30分後から90分間、ラットの自発運動量をSUPERMEX装置(室町機械)により測定した。
Example 8:
Examination of central side effects of tipepidine, or a combination of tipepidine and a CYP inhibitor, SD male rats (7 weeks old) were forcibly orally administered with a solvent or the CYP inhibitor 1-ABT 100 mg / kg. The vehicle-administered rat group was defined as the solvent-administered group, and the 1-ABT-administered rat group was defined as the 1-ABT-administered group. Two hours after the administration of the solvent or 1-ABT in each group, the solvent or the tipepidine hibenz was 20 mg / kg of the acid salt was administered, and 20 mg / kg of tipepidine hibenzate was forcibly orally administered to the 1-ABT administration group. From 30 minutes after administration of 20 mg / kg of the solvent or tipepidine hibenzate, the spontaneous locomotor activity of the rat was measured by SUPERMEX apparatus (Muromachi Kikai) for 90 minutes.
 結果を図7に示した。チペピジンヒベンズ酸塩20mg/kg単独群、及び1-ABT 100mg/kgとチペピジンヒベンズ酸塩20mg/kg併用群において、溶媒投与群と比較して有意な行動量の上昇は認められなかった。つまり、チペピジンとCYP阻害薬の併用においては、チペピジンの血中濃度が増加し、チペピジンの血中持続性が上昇するが(表4)、中枢性の副作用が認められないことが明らかとなった。この時のチペピジンの血中薬物フリー濃度は2.0nMであり実施例6で中枢性の副作用が認められた予測血中薬物フリー濃度6.4nMと比較して十分低かった。 The results are shown in Figure 7. In the tipepidine hibenzate 20 mg / kg alone group and the combination of 1-ABT 100 mg / kg and tipepidine hibenzate 20 mg / kg groups, no significant increase in the amount of activity was observed as compared with the vehicle administration group. In other words, it was revealed that the combination of tipepidine and the CYP inhibitor increased the blood concentration of tipepidine and increased the blood persistence of tipepidine (Table 4), but no central side effects were observed. .. The blood drug-free concentration of tipepidine at this time was 2.0 nM, which was sufficiently lower than the predicted blood drug-free concentration of 6.4 nM in Example 6 in which central side effects were observed.
実施例9:
チペピジン、又はチペピジン及びCYP阻害薬併用によるチペピジンの抗うつ様作用
 Wistar雄性ラット(9週齢)を試験前日に水泳訓練室に投入して環境に慣らした。その後、水温25±1度の水道水を満たした水槽に動物を投入して、15分間強制水泳訓練試験を行った。水泳後、動物を水槽から取り出し、水滴をふき取りケージに戻した。試験当日、体重測定を行い、水泳訓練室に搬入して環境に慣らした。溶媒又はCYP阻害薬1-ABT 100mg/kgを強制経口投与した。溶媒を投与したラット群を溶媒投与群、1-ABTを投与したラット群を1-ABT投与群とし、その2時間後、溶媒投与群においては溶媒又はチペピジン20mg/kgを、1-ABT投与群においてはチペピジン20mg/kgを強制経口投与を行った。その1時間後、または4時間後に、水温25±1度の水道水を満たした水槽に動物を投入して、チペピジンヒベンズ酸塩投与1時間後、または4時間後に5分間強制水泳訓練試験を行い、前面からビデオカメラで撮影した。無動時間の測定はビデオをブラインドした後、目視にて測定を行った。
Example 9:
Antidepressant-like action of tipepidine by tipepidine or a combination of tipepidine and a CYP inhibitor Wistar male rats (9 weeks old) were placed in a swimming training room on the day before the test to acclimate to the environment. Then, the animal was put into a water tank filled with tap water at a water temperature of 25 ± 1 degree, and a forced swimming training test was conducted for 15 minutes. After swimming, the animals were removed from the aquarium and water droplets were wiped off and returned to their cages. On the day of the test, the body weight was measured and brought into a swimming training room to get accustomed to the environment. Vehicle or CYP inhibitor 1-ABT 100 mg / kg was administered by oral gavage. The solvent-administered rat group was defined as the solvent-administered group, and the 1-ABT-administered rat group was defined as the 1-ABT-administered group. Two hours later, in the solvent-administered group, the solvent or tipepidine 20 mg / kg was administered as the 1-ABT-administered group. In this case, tipepidine 20 mg / kg was forcibly orally administered. One hour later, or four hours later, the animal was placed in a tank filled with tap water at a water temperature of 25 ± 1 ° C., and one hour after the administration of tipepidine hibenzate, or four hours later, a forced swimming training test was conducted for 5 minutes. , Taken from the front with a video camera. The immobility time was measured visually after the video was blinded.
 結果を図8、図9に示した。チペピジンヒベンズ酸塩20mg/kg単独群において、溶媒投与群と比較して有意な無動時間の短縮が認められなかった。一方、CYP阻害薬1-ABT 100mg/kg、及びチペピジンヒベンズ酸塩20mg/kg併用群において、溶媒投与群と比較して有意な無動時間の短縮が認められた。また、この無動時間の短縮が、チペピジンヒベンズ酸塩投与1時間後、および4時間後に認められた。
 実施例4および5において、チペピジンヒベンズ酸塩とCYP阻害薬との併用投与により、血漿中暴露の上昇および半減期の増加が示された。また、実施例8において、チペピジンヒベンズ酸塩とCYP阻害薬の併用投与により、中枢性の副作用が認められないことが示された(図7)。これらの結果から、チペピジンヒベンズ酸塩とCYP阻害薬の併用投与において、チペピジンヒベンズ酸塩単独投与と比較して、中枢性の副作用を発現せず、強い抗うつ様作用を長時間維持することが明らかとなった。また、投与1時間後から強い抗うつ効果を示したことから、即効性がある事が明らかとなった。
The results are shown in FIGS. 8 and 9. In the 20 mg / kg tipepidine hibenzate alone group, no significant reduction in immobility time was observed as compared with the vehicle administration group. On the other hand, in the CYP inhibitor 1-ABT 100 mg / kg and tipepidine hibenzate 20 mg / kg combination group, a significant reduction in immobility time was observed as compared with the solvent administration group. In addition, this reduction in immobility time was observed 1 hour and 4 hours after administration of tipepidine hibenzate.
In Examples 4 and 5, combined administration of tipepidine hibenzate and a CYP inhibitor showed increased plasma exposure and increased half-life. In addition, in Example 8, it was shown that central side effects were not observed by the combined administration of tipepidine hibenzate and a CYP inhibitor (FIG. 7). From these results, in the combined administration of tipepidine hibenzate and CYP inhibitor, compared to the administration of tipepidine hibenzate alone, central side effects are not expressed and a strong antidepressant-like action can be maintained for a long time. It became clear. In addition, since it showed a strong antidepressant effect 1 hour after administration, it was clarified that it has immediate effect.
実施例10: 
Wistar雄性ラット対するチペピジンの覚醒作用
 Wistar雄性ラット(9週齢)を12時間サイクルの明暗室で飼育した。ラットはペントバルビタール50mg/kgの腹腔内投与により麻酔を行い、脳波を測定するため、EEG Electrode WIRESを頭蓋骨に移植し、EMG Recordingのために、背の首あたりにEMG Electrode WIRESを移植した。1週間の回復期間の後、EEG/EMGの記録をPonemah(Data Sciences International, lnc.)を用いて測定を行った。Sleep stageの判定にはSleepsign software(KISSEI COMTEC CO., LTD)を用いた。試験当日、チペピジンヒベンズ酸塩8mg/kg及び80mg/kgを試験開始の30分前に腹腔内投与を行い、その後4時間の睡眠状態をソフトウェアにより判定した。
Example 10:
Awakening Effect of Tipepidine on Wistar Male Rats Wistar male rats (9 weeks old) were bred in a dark room with a 12-hour cycle. Rats were anesthetized by intraperitoneal administration of pentobarbital 50 mg / kg, and EEG Electrode WIRES was transplanted to the skull for measuring EEG, and EMG Electrode WIRES was transplanted around the back neck for EMG Recording. After a one week recovery period, EEG / EMG recordings were measured using Ponemah (Data Sciences International, Inc.). The Sleep stage software (KISSEI COMTEC CO., LTD) was used for the determination of the sleep stage. On the day of the test, tipepidine hibenzate 8 mg / kg and 80 mg / kg were intraperitoneally administered 30 minutes before the start of the test, and the sleep state for 4 hours thereafter was determined by software.
 結果を図10A、10B、11Aおよび11Bに示した。チペピジンヒベンズ酸塩8mg/kg及び80mg/kg投与により、溶媒投与群に比較して覚醒時間を維持した。さらに、8mg/kg及び80mg/kg投与においてレム睡眠時間の短縮が認められた。つまり、チペピジンは強い覚醒作用を有していることが明らかとなった。 The results are shown in FIGS. 10A, 10B, 11A and 11B. The administration of tipepidine hibenzate 8 mg / kg and 80 mg / kg maintained the wake time as compared with the vehicle administration group. Furthermore, shortening of REM sleep time was observed at 8 mg / kg and 80 mg / kg administration. That is, it was revealed that tipepidine has a strong wakefulness effect.
実施例11:
SDラットに対するチペピジンの慢性疲労症候群改善作用
 SDラットに、23度に保温した水を1.5cmの高さで張った飼育箱で3日間飼育し、睡眠断眠に伴う慢性疲労症候群モデルを作成した。そのラットに対して、溶媒及びチペピジンクエン酸塩1mg/kg又は5mg/kgの用量で皮下投与を行い、その60分後に強制水泳試験を実施した。
Example 11:
Effect of tipepidine for improving chronic fatigue syndrome on SD rats SD rats were bred for 3 days in a breeding box in which water kept at 23 degrees was placed at a height of 1.5 cm to prepare a model of chronic fatigue syndrome associated with sleep deprivation. .. The rats were subcutaneously administered with a solvent and tipepidine citrate at a dose of 1 mg / kg or 5 mg / kg, and 60 minutes after that, a forced swimming test was performed.
 結果を図12に示した。チペピジンクエン酸塩1mg/kg、5mg/kg群において、溶媒投与群と比較して有意な無動時間の短縮が認められた。 The results are shown in Figure 12. A significant reduction in immobility time was observed in the tipepidine citrate 1 mg / kg and 5 mg / kg groups as compared with the solvent administration group.
実施例12:
チペピジン及びチペピジンとCYP阻害薬併用によるWistarラットに対するチペピジンの覚醒作用
 Wistar雄性ラット(9週齢)を12時間サイクルの明暗室で飼育した。ラットはペントバルビタール50mg/kgの腹腔内投与により麻酔を行い、脳波を測定するため、EEG Electrode WIRESを頭蓋骨に移植し、EMG Recordingのために、背の首あたりにEMG Electrode WIRESを移植する。1週間の回復期間の後、EEG/EMGの記録をPonemah(Data Sciences International, lnc.)を用いて測定を行った。Sleep stageの判定にはSleepsign software(KISSEI COMTEC CO., LTD)を用いた。試験前日、溶媒またはCYP阻害薬1-ABT 100mg/kgを強制経口投与した。溶媒を投与したラット群を溶媒前日投与群、1-ABTを投与したラット群を1-ABT前日投与群とし、溶媒前日投与群及び1-ABT前日投与群それぞれに溶媒又はチペピジン40mg/kgを試験開始の15分前に強制経口投与し、その後6時間の睡眠状態をソフトウェアにより判定した。
 結果を図13A、13B、14Aおよび14Bに示した。チペピジンヒベンズ酸塩40mg/kg単独投与群において、溶媒投与群と比較して有意な覚醒時間の維持及びレム睡眠時間の短縮が認められなかった。一方、CYP阻害薬1-ABT 100mg/kg及びチペピジンヒベンズ酸塩40mg/kg投与群において、1-ABT 100mg/kg及び溶媒投与群と比較して有意な覚醒時間の維持及びレム睡眠時間の短縮が認められた。この結果から、チペピジンヒベンズ酸塩とCYP阻害薬の併用投与において、チペピジンヒベンズ酸塩単独投与と比較して、強い覚醒作用を長時間維持することが明らかとなった。
Example 12:
Awakening Effect of Tipepidine on Wistar Rats by Tipepidine and a Combination of Tipepidine and CYP Inhibitor Wistar male rats (9 weeks old) were bred in a light-dark room with a 12-hour cycle. Rats are anesthetized by intraperitoneal administration of pentobarbital 50 mg / kg, and EEG Electrode WIRES is transplanted to the skull to measure EEG, and EMG Electrode WIRES is transplanted around the back neck for EMG Recording. After a one week recovery period, EEG / EMG recordings were measured using Ponemah (Data Sciences International, Inc.). The Sleep stage software (KISSEI COMTEC CO., LTD) was used for the determination of the sleep stage. The day before the test, 100 mg / kg of vehicle or CYP inhibitor 1-ABT was orally administered by gavage. The vehicle-administered rat group was defined as the solvent-previous day administration group, the 1-ABT-administered rat group was defined as the 1-ABT previous-day administration group, and the solvent-previous day administration group and the 1-ABT previous-day administration group were tested with solvent or tipepidine 40 mg / kg Gavage was administered 15 minutes before the start, and the sleep state for 6 hours thereafter was determined by software.
The results are shown in Figures 13A, 13B, 14A and 14B. In the tipepidine hibenzate 40 mg / kg single administration group, significant maintenance of wakefulness and shortening of REM sleep time were not observed as compared with the vehicle administration group. On the other hand, in the CYP inhibitor 1-ABT 100 mg / kg and tipepidine hibenzate 40 mg / kg administration groups, significant awakening time maintenance and shortening of REM sleep time were observed as compared with 1-ABT 100 mg / kg and vehicle administration groups. Admitted. From these results, it was revealed that the combined administration of tipepidine hibenzate and the CYP inhibitor maintains a strong wakefulness for a long time, as compared with the administration of tipepidine hibenzate alone.
実施例13:
チペピジン、又はチペピジン及びCYP阻害薬併用によるSDラットに対する慢性疲労症候群改善作用
 SDラットを23度に保温した水を1.5cmの高さで張った飼育箱で3日間飼育し、睡眠断眠に伴う慢性疲労症候群モデルを作成する。試験当日、溶媒またはCYP阻害薬をチペピジンまたはその薬学的に許容される塩投与1時間前に強制経口投与し、チペピジンまたはその薬学的に許容される塩を試験開始の1時間前に皮下投与を行う。その60分後に強制水泳試験を実施する。
Example 13:
Amelioration of chronic fatigue syndrome on SD rats by tipepidine or a combination of tipepidine and a CYP inhibitor. SD rats were bred for 3 days in a breeding box with water kept at 23 degrees and a height of 1.5 cm. Create a chronic fatigue syndrome model. On the day of the test, vehicle or CYP inhibitor was administered by oral gavage 1 hour before the administration of tipepidine or a pharmaceutically acceptable salt thereof, and subcutaneous administration of tipepidine or a pharmaceutically acceptable salt thereof was conducted 1 hour before the start of the test. To do. A forced swimming test is carried out 60 minutes later.
 実施例14:
ナルコレプシーマウスに対するチペピジンの覚醒作用及びカタプレキシー抑制作用
 ナルコレプシーマウスに、溶媒及びチペピジンまたはその製薬学的に許容される塩を腹腔内もしくは皮下投与を行い、その後数時間脳波、筋電図及び行動をビデオ撮影する。脳波から覚醒時間を算出し、また脳波筋電図及びビデオ撮影からカタプレキシーの生じる頻度を測定する。
Example 14:
Awakening effect and cataplexic inhibitory effect of tipepidine on narcolepsy mouse Intraperitoneal or subcutaneous administration of solvent and tipepidine or a pharmaceutically acceptable salt thereof to narcolepsy mouse, and then videographed electroencephalogram, electromyogram and behavior for several hours. To do. Awakening time is calculated from EEG, and the frequency of cataplexy is measured from EEG electromyography and video recording.
 実施例15:
チペピジン、又はチペピジン及びCYP阻害薬併用によるナルコレプシーマウスに対する覚醒作用及びカタプレキシー抑制作用
 ナルコレプシーマウスに、溶媒またはCYP阻害薬をチペピジンまたはその薬学的に許容される塩投与1時間前に強制経口投与し、その後チペピジンまたはその薬学的に許容される塩の強制経口投与を行う。その後数時間脳波、筋電図及び行動をビデオ撮影する。脳波から覚醒時間を算出し、また脳波筋電図及びビデオ撮影からカタプレキシーの生じる頻度を測定する。
Example 15:
Awakening action and cataplexy inhibitory action on narcolepsy mouse by tipepidine or a combination of tipepidine and CYP inhibitor , a solvent or CYP inhibitor is forcibly orally administered 1 hour before administration of a solvent or a CYP inhibitor to tipepidine or a pharmaceutically acceptable salt thereof. The oral administration of tipepidine or a pharmaceutically acceptable salt thereof is performed by oral gavage. After that, for several hours, videographs of EEG, EMG, and behavior will be taken. Awakening time is calculated from EEG, and the frequency of cataplexy is measured from EEG electromyography and video recording.
実施例16: 
チペピジン及びCYP2D6阻害薬併用によるナルコレプシー患者における覚醒作用の評価
 ナルコレプシー患者は日中の過度の眠気、情動脱力発作、睡眠麻痺等を示し、日本における有病率は0.16%程度であると報告されている。日中の過度の眠気に対してモダフィニルが有効性を示すが、不応答の患者が20%程度いることが報告されており、さらなる薬剤が求められている。
 ナルコレプシー患者に対してCYP2D6阻害薬(1-300mg)を投薬し、さらに1日当たり1~3回チペピジンまたはその製薬学的に許容される塩(20-200mg)を投薬する。その後、2時間ごとに4セクション20分間の睡眠維持検査を行い、睡眠に至るまでの睡眠潜時を測定する。
Example 16:
Evaluation of wakefulness in narcolepsy patients by combined use of tipepidine and CYP2D6 inhibitors Narcolepsy patients exhibit excessive daytime sleepiness, emotional weakness, paralysis of sleep, etc., and the prevalence in Japan is reported to be about 0.16%. ing. Although modafinil is effective against excessive daytime sleepiness, it has been reported that about 20% of patients do not respond, and further drugs are required.
Patients with narcolepsy are dosed with CYP2D6 inhibitor (1-300 mg) and 1-3 times daily with tipepidine or a pharmaceutically acceptable salt thereof (20-200 mg). Thereafter, a sleep maintenance test for 4 sections and 20 minutes is performed every 2 hours, and sleep latency until sleep is measured.
実施例17:
チペピジン及びCYP2D6阻害薬併用によるナルコレプシー患者における情動脱力発作評価
 ナルコレプシー患者は喜怒哀楽等の過度の情動刺激により情動脱力発作が生じることが知られている。モダフィニルは情動脱力発作には無効だと報告されている。さらにガンマヒドロキシ酪酸(GBH)が米国ではナルコレプシー患者の情動脱力発作の治療に用いられているが、夜間に2度服薬が必要であり服薬コンプライアンスに大きな課題がある。
 ナルコレプシー患者に対して、CYP2D6阻害薬(1-300mg)とチペピジンまたはその製薬学的に許容される塩(20-200mg)を14日間投薬を行い、14日間の情動脱力発作が行った回数を測定する。
Example 17:
Evaluation of emotional weakness seizures in narcolepsy patients by combination use of tipepidine and CYP2D6 inhibitors It is known that narcolepsy patients suffer from emotional weakness seizures due to excessive emotional stimulation such as emotions. Modafinil is reportedly ineffective for emotional weakness attacks. Further, gamma-hydroxybutyric acid (GBH) is used in the United States for the treatment of emotional weakness attacks in patients with narcolepsy, but it requires two doses at night, which poses a major problem in compliance.
CYP2D6 inhibitor (1-300mg) and tipepidine or its pharmaceutically acceptable salt (20-200mg) were administered to patients with narcolepsy for 14 days, and the number of emotional weakness attacks for 14 days was measured. To do.
実施例18:
チペピジン及びCYP2D6阻害薬併用による睡眠時無呼吸患者における覚醒作用の評価
 睡眠時無呼吸患者は夜間に低呼吸状態になり、夜間の眠りが細分化されるため、熟睡できず、翌日の昼間に強い眠気や倦怠感、集中力の低下が求められる。治療方法として、経鼻的持続陽圧呼吸療法(CPAP法という)を用いて圧をかけた空気を送り込むことで、低呼吸状態を改善させる方法が用いられている。CPAP法により改善が認められない場合は、モダフィニル等により日中の過度の眠気を抑制させるが、その作用は十分ではないことが報告されている。
 睡眠時無呼吸患者に対してCYP2D6阻害薬(1-200mg)を投与し、さらに1日当たり1~3回チペピジンまたはその製薬学的に許容される塩(20-200mg)を投与する。その後、20分間を2時間ごとに4セクション睡眠維持検査を行い、睡眠潜時を測定する。
Example 18:
Evaluation of wakefulness in sleep apnea patients by combined use of tipepidine and CYP2D6 inhibitors Sleep apnea patients become hypopnea at night, and sleep at night is subdivided so that they cannot sleep deeply and are strong in the daytime of the next day. Drowsiness, malaise, and poor concentration are required. As a treatment method, there is used a method of improving a hypopnea state by sending pressured air using nasal continuous positive pressure breathing therapy (referred to as CPAP method). When no improvement is observed by the CPAP method, modafinil or the like suppresses excessive daytime sleepiness, but it is reported that its action is not sufficient.
A sleep apnea patient is administered a CYP2D6 inhibitor (1-200 mg), and is further administered 1-3 times daily with tipepidine or a pharmaceutically acceptable salt thereof (20-200 mg). Then, for 20 minutes, a 4-section sleep maintenance test is performed every 2 hours to measure sleep latency.
実施例19: 
チペピジン及びCYP2D6阻害薬併用によるパーキンソン病患者における覚醒作用の評価
 パーキンソン病患者の約80%が睡眠障害を生じていると言われており、非運動症状として最も頻度が多い症状である。特にパーキンソン病患者では昼間に強い眠気が認められ、患者のQuality of lifeを大きく引き下げる要因の一つとして知られている。日中の過度の眠気に対しては適応外処方ではあるが、モダフィニルの使用が推奨されている。しかしながら、モダフィニルの有効性については十分な検証がなされていない。
 パーキンソン病患者に対してCYP2D6阻害薬(1-300mg)を投与し、さらに1日当たり1~3回チペピジンまたはその製薬学的に許容される塩(20-200mg)を投与する。その後、20分間を2時間ごとに4セクション睡眠維持検査を行い、睡眠潜時を測定する。
Example 19:
Evaluation of wakefulness effect in Parkinson's disease patients by combined use of tipepidine and CYP2D6 inhibitors It is said that about 80% of Parkinson's disease patients have sleep disorders, which is the most frequent non-motor symptom. Particularly in patients with Parkinson's disease, strong drowsiness is observed in the daytime, which is known as one of the factors that significantly reduce the quality of life of patients. Although off-label for excessive daytime sleepiness, modafinil is recommended for use. However, the efficacy of modafinil has not been fully verified.
CYP2D6 inhibitor (1-300 mg) is administered to patients with Parkinson's disease, and further tipepidine or a pharmaceutically acceptable salt thereof (20-200 mg) is administered 1 to 3 times a day. Then, for 20 minutes, a 4-section sleep maintenance test is performed every 2 hours to measure sleep latency.
 実施例20:
ガラス玉覆い隠し試験を用いたマウスに対するチペピジンの強迫症改善作用
 ケージに床敷きを敷き、その上にガラス玉を1ケージあたり20個置く。マウスに、溶媒及びチペピジンまたはその製薬学的に許容される塩を腹腔内もしくは皮下投与を行う。投与後のマウスをケージ内に置き、15分間自由行動をさせ、ガラス玉を埋めた個数を目視により計測する。
Example 20:
Obsessive-compulsive effect of tipepidine for mice using the marble burying test. A cage is covered with bedding, and 20 marbles are placed on each cage. The mice are intraperitoneally or subcutaneously administered with a solvent and tipepidine or a pharmaceutically acceptable salt thereof. After administration, the mouse is placed in a cage, allowed to move freely for 15 minutes, and the number of filled glass beads is counted visually.
 実施例21:
ガラス玉覆い隠し試験を用いたチペピジン、又はチペピジン及びCYP阻害薬併用によるSDラットに対する強迫症改善作用
 ケージに床敷きを敷き、その上にガラス玉を1ケージあたり20個置く。マウスに溶媒またはCYP阻害薬を強制経口投与し、その1時間後にチペピジンまたはその薬学的に許容される塩の強制経口投与を行う。投与後のマウスをケージ内に置き、15分間自由行動をさせ、ガラス玉を埋めた個数を目視により計測する。
Example 21:
Obsessive-compulsive amelioration effect on SD rats by tipepidine or a combination of tipepidine and a CYP inhibitor using a glass bead masking test. A cage is covered with bedding, and 20 glass beads are placed on each cage. The vehicle or the CYP inhibitor is orally administered by gavage to the mouse, and 1 hour after that, tipepidine or a pharmaceutically acceptable salt thereof is orally administered by gavage. After administration, the mouse is placed in a cage, allowed to move freely for 15 minutes, and the number of filled glass beads is counted visually.
 実施例22:
恐怖条件付け試験を用いたラットに対するチペピジンの心的外傷後ストレス障害改善作用
 ラットをチャンバー内にいれ、床グリッドから電気ショックを提示し、条件付けを実施する。条件付けの一定時間(24時間)後、ラットに、溶媒及びチペピジンまたはその製薬学的に許容される塩を腹腔内もしくは皮下投与を行う。投与後のラットを条件付けチャンバーにいれ、電気刺激のない条件でのすくみ行動の割合を測定する。
Example 22:
Post-traumatic stress disorder improving effect of tipepidine on rats using fear conditioning test Rats are placed in a chamber, and electric shock is presented from the floor grid to perform conditioning. After a fixed time (24 hours) of conditioning, the rats are intraperitoneally or subcutaneously administered with a solvent and tipepidine or a pharmaceutically acceptable salt thereof. After administration, the rat is placed in a conditioning chamber and the rate of freezing behavior in the absence of electrical stimulation is measured.
 実施例23:
恐怖条件付け試験を用いたチペピジン、又はチペピジン及びCYP阻害薬併用によるSDラットに対する心的外傷後ストレス障害改善作用
 ラットをチャンバー内にいれ、床グリッドから電気ショックを提示し、条件付けを実施する。条件付けの一定時間(24時間)後、ラットに溶媒またはCYP阻害薬を強制経口投与し、その1時間後にチペピジンまたはその薬学的に許容される塩の強制経口投与を行う。投与後のラットを条件付けチャンバーにいれ、電気刺激のない条件でのすくみ行動の割合を測定する。
Example 23:
Post-traumatic stress disorder ameliorative effect on SD rats by tipepidine or a combination of tipepidine and a CYP inhibitor using a fear conditioning test. Rats are placed in a chamber, and electric shock is presented from the floor grid to perform conditioning. After a certain period of conditioning (24 hours), the rats are gavaged with solvent or CYP inhibitor, and 1 hour after that, gipipidine or a pharmaceutically acceptable salt thereof is gavaged. After administration, the rat is placed in a conditioning chamber and the rate of freezing behavior in the absence of electrical stimulation is measured.
 本発明は、医薬品などの分野、例えば、中枢性疾患の治療薬及び予防薬の開発、又は製造分野において利用可能である。 The present invention can be used in the fields of pharmaceuticals and the like, for example, in the fields of development or production of therapeutic and prophylactic agents for central diseases.

Claims (33)

  1.  CYP2D6阻害薬と併用される、チペピジンまたはその製薬学的に許容される塩を含む、中枢性疾患を治療または予防するための医薬組成物。 A pharmaceutical composition for treating or preventing a central disease, which comprises tipepidine or a pharmaceutically acceptable salt thereof, which is used in combination with a CYP2D6 inhibitor.
  2.  チペピジンまたはその製薬学的に許容される塩と併用される、CYP2D6阻害薬を含む、中枢性疾患を治療または予防するための医薬組成物。 A pharmaceutical composition for treating or preventing a central disease, which comprises a CYP2D6 inhibitor in combination with tipepidine or a pharmaceutically acceptable salt thereof.
  3.  チペピジンまたはその製薬学的に許容される塩およびCYP2D6阻害薬を含む、中枢性疾患を治療または予防するための医薬組成物。 A pharmaceutical composition for treating or preventing a central disease, which comprises tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
  4.  CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブ、デュロキセチン、ミラベグロン、セレコキシブ、エスシタロプラム、モクロベミド、シメチジン、フルボキサミン、アビラテロン、アミオダロン、クロバザム、コビシスタット、デスベンラファキシン、ラベタロール、ロルカセリン、リトナビル、セルトラリン、ベムラフェニブ、およびデラムシクランからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、請求項1~3のいずれか一項に記載の医薬組成物。 CYP2D6 inhibitors are quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, duloxetine, mirabegron, celecoxib, escitalopram, moclobelatil, clavitone, fluvoxamine, abiraterabalone, mirabedalone, amiodarone, fluvoxamine, abiraterabalone, amiodalone, amiodala. The pharmaceutical composition according to any one of claims 1 to 3, which is one or more drugs selected from the group consisting of, ritonavir, sertraline, vemurafenib, and deramciclane, or a pharmaceutically acceptable salt thereof. ..
  5.  CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、ダコミチニブ、デュロキセチン、ミラベグロン、セレコキシブ、エスシタロプラム、モクロベミド、シメチジン、およびフルボキサミンからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、請求項1~3のいずれか一項に記載の医薬組成物。 The CYP2D6 inhibitor is one or more pharmaceutical agents selected from the group consisting of quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, dacomitinib, duloxetine, mirabegron, celecoxib, escitalopram, moclobemide, cimetidine, and fluvoxamine; The pharmaceutical composition according to any one of claims 1 to 3, wherein the pharmaceutical composition is an acceptable salt.
  6.  CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、フルオキセチン、テルビナフィン、シナカルセト、およびダコミチニブからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、請求項1~3のいずれか一項に記載の医薬組成物。 The CYP2D6 inhibitor is one or more drugs selected from the group consisting of quinidine, paroxetine, bupropion, fluoxetine, terbinafine, cinacalcet, and dacomitinib, or a pharmaceutically acceptable salt thereof. The pharmaceutical composition according to any one of claims.
  7.  CYP2D6阻害薬が、キニジン、パロキセチン、ブプロピオン、およびフルオキセチンからなる群から選択される一種以上の薬剤、またはその製薬学的に許容される塩である、請求項1~3のいずれか一項に記載の医薬組成物。 4. The CYP2D6 inhibitor is one or more agents selected from the group consisting of quinidine, paroxetine, bupropion, and fluoxetine, or a pharmaceutically acceptable salt thereof. Pharmaceutical composition.
  8.  CYP2D6阻害薬が、キニジン、またはその製薬学的に許容される塩である、請求項1~3のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 3, wherein the CYP2D6 inhibitor is quinidine or a pharmaceutically acceptable salt thereof.
  9.  中枢性疾患が、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症である、請求項1~8のいずれか一項に記載の医薬組成物。 Central diseases are Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorders, chronic fatigue syndrome, fatigue associated with neurodegenerative and central diseases, overeating, addiction, or fibromyalgia, The pharmaceutical composition according to any one of 1 to 8.
  10.  中枢性疾患が、睡眠障害である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is sleep disorder.
  11.  中枢性疾患が、過眠、特発性過眠障害、またはナルコレプシーである、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is hypersomnia, idiopathic hypersomnia disorder, or narcolepsy.
  12.  中枢性疾患が、抑うつ障害群である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is a depressive disorder group.
  13.  中枢性疾患が、不安性の苦痛を伴う抑うつ障害群、混合性の特徴を伴う抑うつ障害群、メランコリアの特徴を伴う抑うつ障害群、気分に一致する精神病性の特徴を伴う抑うつ障害群、気分に一致しない精神病性の特徴を伴う抑うつ障害群、緊張病を伴う抑うつ障害群、季節型抑うつ障害群、重篤気分調整症、うつ病、持続性抑うつ障害、月経前不快気分障害、または物質・医薬品誘発性抑うつ障害である、請求項1~8のいずれか一項に記載の医薬組成物。 Central disorders include anxiety-related depressive disorders, depressive disorders with mixed features, depressive disorders with melancholic features, depressive disorders with mood-matching psychotic features, mood Depressive disorders with inconsistent psychotic features, depressive disorders with catatonic disorders, seasonal depressive disorders, severe dysregulation, depression, persistent depressive disorder, premenstrual dysphoric disorder, or substances / medicines The pharmaceutical composition according to any one of claims 1 to 8, which is an induced depressive disorder.
  14.  中枢性疾患が、小児における抑うつ障害群である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is a depressive disorder group in children.
  15.  中枢性疾患が、小児における不安性の苦痛を伴う抑うつ障害群、小児における混合性の特徴を伴う抑うつ障害群、小児におけるメランコリアの特徴を伴う抑うつ障害群、小児における気分に一致する精神病性の特徴を伴う抑うつ障害群、小児における気分に一致しない精神病性の特徴を伴う抑うつ障害群、小児における緊張病を伴う抑うつ障害群、小児における季節型抑うつ障害群、小児における重篤気分調整症、小児におけるうつ病、小児における持続性抑うつ障害、小児における月経前不快気分障害、または小児における物質・医薬品誘発性抑うつ障害である、請求項1~8のいずれか一項に記載の医薬組成物。 Central disorders are anxiety-affected depressive disorders in children, depressive disorders with mixed features in children, depressive disorders with melancholic features in children, psychotic features that match mood in children Depressive disorder group with illness, depressive disorder group with psychotic features inconsistent with mood in children, depressive disorder group with catatonic disorders in children, seasonal depressive disorder group in children, severe mood regulation in children, in children The pharmaceutical composition according to any one of claims 1 to 8, which is depression, persistent depressive disorder in children, premenstrual dysphoric disorder in children, or substance / medicine-induced depressive disorder in children.
  16.  中枢性疾患が、不安症群である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is an anxiety group.
  17.  中枢性疾患が、分離不安症、限局性恐怖症、社交不安症、パニック症、または物質・医薬品誘発性不安症である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is separation anxiety disorder, localized phobia, social anxiety disorder, panic disorder, or substance / drug-induced anxiety disorder.
  18.  中枢性疾患が、小児における不安症群である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is an anxiety group in children.
  19.  中枢性疾患が、小児における分離不安症、小児における限局性恐怖症、小児における社交不安症、小児におけるパニック症、または小児における物質・医薬品誘発性不安症である、請求項1~8のいずれか一項に記載の医薬組成物。 9. The central illness is segregated anxiety in children, localized phobia in children, social anxiety in children, panic disorder in children, or substance / drug-induced anxiety in children. The pharmaceutical composition according to one paragraph.
  20.  中枢性疾患が、神経発達症群である、請求項1~8のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 8, wherein the central disease is a neurodevelopmental group.
  21.  中枢性疾患が、自閉症スペクトラム症、注意欠如・多動症、限局性学習症、知的能力障害、言語症、語音症、小児期発症流暢症、社会的(語用論的)コミュニケーション症、発達性協調運動症、またはチック症群である、請求項1~8のいずれか一項に記載の医薬組成物。 Central diseases include autism spectrum disorder, attention deficit / hyperactivity disorder, localized learning disorder, intellectual disability, speech disorders, speech disorders, childhood-onset fluency, social (pragmatic) communication disorders, development The pharmaceutical composition according to any one of claims 1 to 8, which is a group having sexual dyskinesia or tic disorder.
  22.  チペピジンまたはその製薬学的に許容される塩の投与量が一日当たり2mg~2000mgである、請求項1~21のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 21, wherein the dose of tipepidine or a pharmaceutically acceptable salt thereof is 2 mg to 2000 mg per day.
  23.  CYP2D6阻害薬の投与量が一日当たり1mg~1000mgである、請求項1~22のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 22, wherein the dose of the CYP2D6 inhibitor is 1 mg to 1000 mg per day.
  24.  治療上の有効量のチペピジンまたはその製薬学的に許容される塩と治療上の有効量のCYP2D6阻害薬を哺乳動物に投与することを含む、中枢性疾患を治療または予防するための方法。 A method for treating or preventing a central disease, which comprises administering to a mammal a therapeutically effective amount of tipepidine or a pharmaceutically acceptable salt thereof and a therapeutically effective amount of a CYP2D6 inhibitor.
  25.  CYP2D6阻害薬と併用される、中枢性疾患の治療または予防における使用のための、チペピジンまたはその製薬学的に許容される塩。 Tipepidine or a pharmaceutically acceptable salt thereof for use in the treatment or prevention of central diseases, which is used in combination with a CYP2D6 inhibitor.
  26.  チペピジンまたはその製薬学的に許容される塩と併用される、中枢性疾患の治療または予防における使用のための、CYP2D6阻害薬。 A CYP2D6 inhibitor for use in the treatment or prevention of central diseases, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
  27.  CYP2D6阻害薬と併用される、中枢性疾患を治療または予防するための医薬の製造のための、チペピジンまたはその製薬学的に許容される塩の使用。 Use of tipepidine or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing central diseases, which is used in combination with a CYP2D6 inhibitor.
  28.  チペピジンまたはその製薬学的に許容される塩と併用される、中枢性疾患を治療または予防するための医薬の製造のための、CYP2D6阻害薬の使用。 Use of a CYP2D6 inhibitor for the manufacture of a medicament for treating or preventing central diseases, which is used in combination with tipepidine or a pharmaceutically acceptable salt thereof.
  29.  中枢性疾患を治療または予防するための医薬の製造のための、チペピジンまたはその製薬学的に許容される塩およびCYP2D6阻害薬の使用。 Use of tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor for the manufacture of a medicament for treating or preventing a central disease.
  30.  チペピジンまたはその製薬学的に許容される塩と、CYP2D6阻害薬を含む、中枢性疾患の治療または予防のためのキット。 A kit for the treatment or prevention of central diseases, which contains tipepidine or a pharmaceutically acceptable salt thereof and a CYP2D6 inhibitor.
  31.  チペピジンまたはその製薬学的に許容される塩を含む、パーキンソン病、抗精神病薬使用によるパーキンソン病症候群、睡眠障害、慢性疲労症候群、神経変性及び中枢性疾患に伴う疲労感、過食、依存症、または線維筋痛症の治療剤または予防剤。 Including tipepidine or a pharmaceutically acceptable salt thereof, Parkinson's disease, Parkinson's disease syndrome due to antipsychotic use, sleep disorder, chronic fatigue syndrome, fatigue associated with neurodegeneration and central diseases, overeating, addiction, or A therapeutic or preventive agent for fibromyalgia.
  32.  疾患が、睡眠障害である、請求項31に記載の治療剤または予防剤。 The therapeutic or prophylactic agent according to claim 31, wherein the disease is sleep disorder.
  33.  疾患が、過眠、特発性過眠障害、またはナルコレプシーである、請求項31に記載の治療剤または予防剤。 The therapeutic or prophylactic agent according to claim 31, wherein the disease is hypersomnia, idiopathic hypersomnia disorder, or narcolepsy.
PCT/JP2019/043606 2018-11-08 2019-11-07 Therapeutic agent for central nervous system disease including tipepidine WO2020095979A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2018-210469 2018-11-08
JP2018210469 2018-11-08
JP2019148875 2019-08-14
JP2019-148875 2019-08-14

Publications (1)

Publication Number Publication Date
WO2020095979A1 true WO2020095979A1 (en) 2020-05-14

Family

ID=70612044

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/043606 WO2020095979A1 (en) 2018-11-08 2019-11-07 Therapeutic agent for central nervous system disease including tipepidine

Country Status (1)

Country Link
WO (1) WO2020095979A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003523936A (en) * 1999-04-07 2003-08-12 ファイザー・プロダクツ・インク Use of CYP2D6 inhibitors in combination therapy
JP2005501070A (en) * 2001-08-08 2005-01-13 イーライ・リリー・アンド・カンパニー Combination therapy to treat neurological disorders
JP2009227631A (en) * 2008-03-25 2009-10-08 Kumamoto Univ Therapeutic agent for mood disturbance or emotional disturbance
JP2011246446A (en) * 2010-04-28 2011-12-08 Kumamoto Univ Therapeutic agent for brain dysfunction
JP2013063958A (en) * 2011-09-01 2013-04-11 Kumamoto Univ Therapeutic agent for schizophrenia

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003523936A (en) * 1999-04-07 2003-08-12 ファイザー・プロダクツ・インク Use of CYP2D6 inhibitors in combination therapy
JP2005501070A (en) * 2001-08-08 2005-01-13 イーライ・リリー・アンド・カンパニー Combination therapy to treat neurological disorders
JP2009227631A (en) * 2008-03-25 2009-10-08 Kumamoto Univ Therapeutic agent for mood disturbance or emotional disturbance
JP2011246446A (en) * 2010-04-28 2011-12-08 Kumamoto Univ Therapeutic agent for brain dysfunction
JP2013063958A (en) * 2011-09-01 2013-04-11 Kumamoto Univ Therapeutic agent for schizophrenia

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HIRIKAWA, HIDEO: "Organic medicine molecular theory -Chemical structure, pharmacological activity and drug discovery", KYOTO HIROKADA PUBLISHING INC., article "Inhibition and induction of metabolic enzymes", pages: 186 - 188 *
IWASE, MARIKO: "The appropriate use of OTC drugs in multiple medication", FINAL RESEARCH REPORT ON GRANTS-IN-AID FOR SCIENTIFIC RESEARCH, 29 May 2015 (2015-05-29) *

Similar Documents

Publication Publication Date Title
JP6927601B2 (en) Compositions and Methods Containing Bupropion or Related Compositions and Dextromethorphan
JP7155206B2 (en) VMAT2 inhibitors for treating neurological diseases or disorders
JP6842129B2 (en) Compositions and Methods Containing Bupropion or Related Compositions and Dextromethorphan
EP3463323B1 (en) Solid oral dosage forms of 2r,6r-hydroxynorketamine or derivatives thereof
JP2020023543A6 (en) Compositions and methods comprising bupropion or related compositions and dextromethorphan
AU2008281112A1 (en) Use of KCNQ potassium channel openers for reducing symptoms of or treating disorders or conditions wherein the dopaminergic system is disrupted
US20220096444A1 (en) Prophylactic or therapeutic agent for delirium
JP2016053095A (en) Methods and compositions for treating depression using cyclobenzaprine
US20080242682A1 (en) Preventive or Therapeutic Agent for Sleep Disorder
TW202327571A (en) Psychoactive medicines and their use for treating psychiatric and neurological conditions and disorders
JP2022519541A (en) Compositions and Methods for Treating Anxiety-Related Disorders
WO2020095979A1 (en) Therapeutic agent for central nervous system disease including tipepidine
US20230181521A1 (en) Isotopically enriched analogs of 5,6-methylenedioxy-2-aminoindane (mdai)
TW202345789A (en) Psychoactive medicines and their use for treating psychiatric and neurological conditions and disorders
BR112017027839B1 (en) USE OF VMAT2 INHIBITORS FOR PREPARATION OF A PHARMACEUTICAL COMPOSITION TO TREAT MANIA IN A MOOD DISORDER IN AN INDIVIDUAL; AND/OR TREAT OBSESSIVE-COMPULSIVE DISORDER (OCD) REFRACTORY TO TREATMENT
JP2021017412A (en) Pharmaceutical combination of tipepidine and cyp2d6 inhibitor
US20090203607A1 (en) Method And Compositions For Simultaneously Regulating Memory And Mood
US20070004724A1 (en) (+)-(2S,3s)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinor for treating anxiety
US20220378764A1 (en) Modulating expression level of a gene encoding a cytochrome p450 protein by treating a human subject with a nitroxide
NZ614725B2 (en) Methods and compositions for treating depression using cyclobenzaprine

Legal Events

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

Ref document number: 19881417

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19881417

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP