US20170326127A1 - Composition and method for treatment of neuropsychiatric disorders - Google Patents

Composition and method for treatment of neuropsychiatric disorders Download PDF

Info

Publication number
US20170326127A1
US20170326127A1 US15/662,289 US201715662289A US2017326127A1 US 20170326127 A1 US20170326127 A1 US 20170326127A1 US 201715662289 A US201715662289 A US 201715662289A US 2017326127 A1 US2017326127 A1 US 2017326127A1
Authority
US
United States
Prior art keywords
disorder
component
combination therapy
disorders
therapy composition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/662,289
Other languages
English (en)
Inventor
Tong Hyon LEE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Duke University
Original Assignee
Generys Biopharmaceuticals Corp
Generys Korea Corp
Duke University
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 Generys Biopharmaceuticals Corp, Generys Korea Corp, Duke University filed Critical Generys Biopharmaceuticals Corp
Priority to US15/662,289 priority Critical patent/US20170326127A1/en
Assigned to DUKE UNIVERSITY, GENERYS KOREA CORPORATION, GENERYS BIOPHARMACEUTICALS CORPORATION reassignment DUKE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEE, Tong Hyon
Publication of US20170326127A1 publication Critical patent/US20170326127A1/en
Assigned to DUKE UNIVERSITY reassignment DUKE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GENERYS BIOPHARMACEUTICALS CORPORATION, GENERYS KOREA CORPORATION
Priority to US16/808,458 priority patent/US20200197383A1/en
Priority to US18/121,626 priority patent/US20230293443A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic 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
    • A61K31/4458Non condensed piperidines, e.g. piperocaine only substituted in position 2, e.g. methylphenidate
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • 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
    • A61K31/48Ergoline derivatives, e.g. lysergic acid, ergotamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention was produced in part using funds from the United States Federal Government under NIH Grant Nos.: R01 DA012768 entitled “Cocaine Withdrawal: A Window of Treatment Opportunity”; R01DA14323 entitled “Medication Development for Methamphetamine Abuse”; NS42124 entitled “Presynaptic Mechanisms and Parkinson's Treatment”; and RC2 DA028905 entitled “Novel Ondansetron Formulation for Combination Treatment of Psychostimulant Abuse.” Accordingly, the Federal Government has certain rights to the present invention.
  • the present invention relates to combination therapy compositions and methods that can temporally modulate underlying dysfunctional neural processes and that can reduce symptoms and/or signs of neuropsychiatric disorders, including, but not limited to, psychostimulant use disorder (PUD) and other substance-related and addictive disorders, post-traumatic stress disorder (PTSD) and other trauma- and stressor-related disorders, and levodopa-induced dyskinesia (LID) and other types of dyskinesia.
  • PID psychostimulant use disorder
  • PTSD post-traumatic stress disorder
  • LID levodopa-induced dyskinesia
  • COC cocaine
  • METH methamphetamine
  • psychostimulants a drug that has a significant comorbid/risk factor in various other neuropsychiatric and medical disorders, e.g., schizophrenia, bipolar disorder, hepatitis C, HIV (See Whiteford et.
  • PUD psychostimulant use disorder
  • Drugs tested in previous clinical trials include other psychostimulants used alone (e.g., d-amphetamine, methylphenidate, etc.), selective dopamine (DA) or serotonin reuptake inhibitors (e.g., fluoxetine), full or partial DA agonists (e.g., aripiprazole), DA antagonists (e.g., risperidone), GABA ( ⁇ -aminobutyric acid) agonists (e.g., gabapentin), and CNS stimulant modafinil.
  • psychostimulants used alone (e.g., d-amphetamine, methylphenidate, etc.), selective dopamine (DA) or serotonin reuptake inhibitors (e.g., fluoxetine), full or partial DA agonists (e.g., aripiprazole), DA antagonists (e.g., risperidone), GABA ( ⁇ -aminobutyric acid) agonist
  • DA agonists and 5HT2, 5-HT3 or NK-1 receptor antagonists can reverse behavioral and neurobiological changes observed in sensitization and self-administration animal models of PUD.
  • 5HT2 5-HT3 or NK-1 receptor antagonists
  • Pharmacologically-mediated reactivation and reconsolidation blockade of the psychostimulant-abuse circuit a novel treatment strategy.
  • Drug and alcohol dependence 124: 11-18 these treatments: (1) reverse previously established COC or METH behavioral sensitization; (2) attenuate self-administration under progressive ratio and drug-induced reinstatement paradigms; and (3) normalize associated neurobiological marker changes.
  • Important findings from these investigations include: (1) consistent treatment efficacy in animal models is achieved if and only if the antagonist is given 3.5 hours after, but not before or with, the agonist; and (2) daily DA agonists or 5-HT antagonists given alone are ineffective.
  • DA replacement treatments are used to alleviate symptoms and signs of PD and other related disorders by restoring DA signaling in the brain.
  • the dopamine (DA) precursor levodopa (L-3,4-dihydroxyphenylalanine) is one of the most effective drugs for alleviating PD symptoms and signs in PD patients.
  • LID levodopa-induced dyskinesia
  • LID is one of the main reasons for the failure of levodopa treatment (with or without adjunctive dopamine agonists) of PD patients.
  • levodopa sensitization plays an important role in the development and maintenance of LID.
  • Dysregulation of DA synthesis, release and clearance of extracellular DA is thought to lead to exaggerated fluctuations in the synaptic DA concentrations following levodopa administration. These fluctuations, in turn, induce dysfunctional synaptic alterations in the basal ganglia and other brain areas.
  • Potential reasons for this less-than-optimal efficacy may include but not limited to: (1) reduced peak levodopa concentrations (for a given “equivalent” dose); (2) development of pharmacodynamic tolerance; and (3) emergence of new, distinct neurobiological alterations during prolonged “continuous” levodopa treatment, a well-documented phenomenon for direct and indirect DA agonists.
  • DBS deep brain stimulation
  • NCS-R National Comorbidity Survey Replication
  • Various neuropsychiatric disorders including PUD, LID and PTSD, may be regarded as a consolidated form of maladaptive dysfunctional neural circuit (dysfunctional neural plasticity).
  • Disorder-specific neural plasticity changes at various levels including but not limited to the levels of neural mechanisms and processes, brain areas, brain area connectivity, brain networks and network interactions within and without the brain, are considered to underlie the signs and symptoms of specific disorders (see Headley and Pare, 2013 (Headley, D B, Pare, D (2013). In sync: gamma oscillations and emotional memory. Front Behav Neurosci 7: 170)).
  • treatment-mediated normalization of such changes in neural plasticity might result in successful therapeutic results.
  • FIG. 1 schematically illustrates the neural processes underlying the maintenance of a dysfunctional neural circuit.
  • the stable, consolidated “PUD neural circuit” may be reactivated by exposure to various stimuli such as psychostimulants themselves (for example, “drug tasting”), environmental cues associated with drug use (e.g., seeing drug paraphernalia) or “stress” (see Childress et al., 1999 (Childress, A R, Mozley, P D, Mcelgin, W, Fitzgerald, J, Reivich, M, O′brien, C P (1999). Limbic activation during cue-induced cocaine craving. The American journal of psychiatry 156: 11-18); Sinha, 2006(Sinha, R, Garcia, M, Paliwal, P, Kreek, M J, Rounsaville, B J (2006).
  • stimuli such as psychostimulants themselves (for example, “drug tasting”), environmental cues associated with drug use (e.g., seeing drug paraphernalia) or “stress” (see Childress et al., 1999 (Childress, A R, Moz
  • LID is another consolidated form of dysfunctional neural plasticity changes (“LID neural circuit”), which is maintained by repeated reactivation following therapeutic levodopa administration and subsequent reconsolidation during pharmacokinetic (PK) and/or pharmacodynamic (PD) decline of therapeutic effects of levodopa.
  • PK pharmacokinetic
  • PD pharmacodynamic
  • the dysfunctional “PTSD neural circuit” in PTSD patients may be conceptualized as being maintained through repeated reactivation and reconsolidation cycles.
  • the dysfunctional neural circuit may be reactivated when a patient is exposed to stimuli that were previously associated with the causative traumatic stressful event (e.g., any loud noise that reminds the patient of a bomb explosion that killed a friend, getting inside a car after being in a major auto accident, etc.). This reactivation leads to reemergence of PTSD symptoms and signs, and symptoms and signs subsides when the reactivated
  • PTSD circuit reconsolidates into the stable but readily-reactivatable state. Upon the reconsolidation, PTSD symptoms and signs abates, ready to reemerge upon the next encounter with stimuli.
  • PID psychostimulant use disorder
  • PTSD post-traumatic stress disorder
  • LID levodopa-induced dyskinesia
  • the present invention provides combination compositions and methods comprised of two active ingredients.
  • One of the ingredients a DA agonist
  • the second ingredient a 5-HT2, 5-HT3 or NK-1 antagonist
  • the invention is designed to sequentially reactivate the dysfunctional neural circuit and to block its reconsolidation, resulting in progressive reduction in the symptoms and/or signs of neuropsychiatric disorders to subclinical levels.
  • disorders that can be treated with the combination therapy include but not limited to psychostimulant use disorder (PUD) and other substance-related and addictive disorders, post-traumatic stress disorder (PTSD) and other trauma- and stressor-related disorders, and levodopa-induced dyskinesia (LID) and other types of dyskinesia.
  • PID psychostimulant use disorder
  • PTSD post-traumatic stress disorder
  • LID levodopa-induced dyskinesia
  • the present invention provides combination therapy compositions and methods, which may, by temporally modulating the underlying dysfunctional neural processes, reduce symptoms and/or signs of neuropsychiatric disorders, including, but not limited to, psychostimulant use disorder (PUD) and other substance-related and addictive disorders, post-traumatic stress disorder (PTSD) and other trauma- and stressor-related disorders, and levodopa-induced dyskinesia (LID) and other types of dyskinesia.
  • PID psychostimulant use disorder
  • PTSD post-traumatic stress disorder
  • LID levodopa-induced dyskinesia
  • compositions and the methods embodied in the present disclosures may be effective in the treatment of: (1) other substance-related disorders, including but not limited to tobacco use disorder, alcohol use disorder and cannabis use disorder; (2) behavioral addictive disorders, including but not limited to compulsive gambling and internet addictions; (3) other trauma and stressor-related disorders, including but not limited to reactive attachment disorder, acute stress disorder and adjustment disorder; (4) anxiety disorders, including but not limited to generalized anxiety disorder, panic disorder, agoraphobia and substance/drug-induced anxiety disorders; (5) obsessive-compulsive disorders, including but not limited to body dysmorphic disorder, hoarding disorder, trichotillomania, and excoriation disorder; and (6) eating disorders, including but not limited to, anorexia nervosa, bulimia nervosa, and binge-eating disorder.
  • other substance-related disorders including but not limited to tobacco use disorder, alcohol use disorder and cannabis use disorder
  • behavioral addictive disorders including but not limited to compulsive gambling and internet addictions
  • FIG. 1 schematically illustrates the neural processes underlying the maintenance of a dysfunctional neural circuit.
  • FIG. 2 shows a conceptual model of combination therapy embodied in the present invention.
  • FIG. 3 shows that a combination of the DA agonist pergolide and the 5-HT2A/2C antagonist ketanserin significantly reduces levodopa-induced abnormal involuntary movement (LIAIM) in rat models of LID.
  • LIAIM levodopa-induced abnormal involuntary movement
  • Target indications described in the present invention may include primary disorders, secondary disorders, side effects, or comorbid conditions.
  • various neuropsychiatric disorders including but not limited to PUD, LID and PTSD, can be conceptualized as stable, consolidated forms of dysfunctional, maladaptive synaptic plasticity, which are maintained by repeated reactivation/reconsolidation cycles. While limited pharmacological options are currently available for various neuropsychiatric disorders, most of these treatments may be considered to be only “palliative” to the extent that they are mainly used to mitigate signs and symptoms of the targeted primary and/or comorbid disorders. There is an unmet medical need for more effective pharmacological treatment that can normalize the underlying dysfunction neural circuit (“disease modification”).
  • FIG. 2 shows a conceptual model of combination therapy embodied in the present invention.
  • the combination therapy is comprised of two active ingredients (Component 1 and Component 2, FIG. 2 ).
  • Component 1 FIG. 1
  • Component 2 incorporated as a therapeutic method, composition, formulation or dosage form into the same embodiment as Component 1 blocks the reconsolidation and subsequent restabilization of the maladaptive memory back to Stable State.
  • the therapeutic embodiments described herein allow for delivery of the combination therapy in optimized and convenient formats and manners, leading to progressive reduction of signs and symptoms of targeted disorders to subclinical levels.
  • the compositions, methods and examples promote patient compliance, and enable the combination therapy to be effectively applied as a single dosage form therapy in clinical practices.
  • the combination therapy may be used for treatment of neuropsychiatric disorders.
  • Disorders that may be treated by the combination therapy include PUD, LID and PTSD.
  • Additional disorders include but not limited to: (1) other substance-related disorders, including but not limited to tobacco use disorder, alcohol use disorder and cannabis use disorder; (2) behavioral addictive disorders, including but not limited to compulsive gambling and internet addictions; (3) other trauma and stressor-related disorders, including but not limited to reactive attachment disorder, acute stress disorder and adjustment disorder; (4) anxiety disorders, including but not limited to generalized anxiety disorder, panic disorder, agoraphobia and substance/drug-induced anxiety disorders; (5) obsessive-compulsive disorders, including but not limited to body dysmorphic disorder, hoarding disorder, trichotillomania, and excoriation disorder; and (6) feeding and eating disorders, including but not limited to anorexia nervosa, bulimia nervosa, and binge-eating disorder.
  • BDNF brain-derived neurotrophic factor
  • AMPA ⁇ -amino-3-hydroxy-5-methyl-4-isoxazolepropionate
  • NMDA N-methyl-D-aspartate
  • the combination therapy may be administered in any compositions, dosage forms, methods or administration routes compatible with clinical practices, which are understood and executed in the field to which the present invention pertains.
  • Component 1 reactivates the maladaptive dysfunctional neural circuit from a stable state to a reactivated and transiently labile state.
  • Component 2 incorporated into the same composition, formulation or dosage form as Component 1 blocks reconsolidation and subsequent restabilization of the reactivated neural circuit to a stable state.
  • the single composition, formulation or dosage form optimizes the combination therapy, and allows the two active ingredients to be delivered in vivo in such a manner to ensure high patient compliance, thereby reducing symptoms and signs of the target disorder to subclinical levels.
  • the compositions, formulations, methods and exemplary embodiments described herein enable the combination therapy to be effectively applied as a single dosage form.
  • Component 1 may be selected from small molecules, other types of molecules, and modifications thereof (for example, formulations, prodrugs, etc.).
  • the other types of molecules include, but are not limited to, small interfering ribonucleic acids (siRNAs), micro-RNAs, antisense oligonucleotides, aptamers, peptides, proteins (e.g., antibodies), naturally occurring organic or inorganic molecules, chemical elements (e.g., lithium), and any synthetic compounds greater or smaller than small molecules.
  • Component 1 may be included in a single composition, formulation and dosage form, together with Component 2, which may be delivered by any safe, acceptable and effective dosing method to reactivate the dysfunctional neural circuit from a stable state to a reactivated and labile state.
  • component 1 is a pharmacologically effective indirect or direct DA agonist or a pharmaceutically acceptable salt thereof, with or without norepinephrine (NE) and/or serotonin (5-HT) agonistic efficacy.
  • NE norepinephrine
  • 5-HT serotonin
  • component 1 is a pharmacologically effective indirect or direct DA agonist or a pharmaceutically acceptable salt thereof, with or without norepinephrine (NE) and/or serotonin (5-HT) agonistic efficacy.
  • NE norepinephrine
  • 5-HT serotonin
  • Component 1 may be also the substances described in Section 1 or modifications thereof, which are or are expected to be functional DA agonists based on: (1) their physiochemical characteristics, as evaluated by any analysis methods known to those skilled in the art, including but not limited to pharmacophore-based “docking”, virtual computational screening and other in vitro/in silico analysis methods; and/or (2) their in vitro and/or in vivo pharmacological profiles.
  • an indirect DA agonist e.g., DA precursor, DA releasing agent or a DA reuptake blocker
  • an agonist that also acts as a pharmacologically effective NE and/or 5-HT agonist e.g., d-amphetamine, methylphenidate, or a selective dopamine/norepinephrine reuptake inhibitor
  • a DA agonist that is not a pharmacologically effective NE and/or 5-HT agonist (e.g., a selective DA reuptake blocker).
  • a direct DA agonist is selected as Component 1
  • a non-selective D1-type or D2-type DA receptor agonist is preferable to a D1-selective or D2-selective receptor agonist.
  • a direct DA agonist is selected as Component 1
  • an agonist that also acts as a pharmacologically effective NE and/or 5-HT agonist is preferable to those showing no NE and/or 5-HT efficacy.
  • Agonists and pharmaceutically-acceptable salts and any modified forms thereof which exhibit immediate and pulsatile release profiles in any in vitro or in vivo tests that are well-known to those skilled in the art (e.g., in vitro dissolution test, direct determination of pharmacokinetic parameters in humans), are preferable to any other combinations/permutations of immediate, delayed, pulsatile, and extended/sustained-release profiles.
  • One preferred in vitro release profile of an agonist is a dissolution rate of greater than or equal to 90% in less than or equal to 2 hours under simulated gastric or intestinal dissolution test conditions, and even more preferred profile is a dissolution rate of greater than or equal to 80% dissolution in less than or equal to 1 hour under identical test conditions.
  • Agonists and pharmaceutically acceptable salts and modifications thereof may have a terminal elimination half life (t1 ⁇ 2) of 20 hours or less in the human body.
  • a preferred terminal elimination half life (t1 ⁇ 2) is 10 hours or less, and an even more preferred terminal elimination half life (t1 ⁇ 2) is 5 hours or less.
  • Component 2 may be selected from small molecules, other types of molecules, and modifications thereof (for example, formulations, prodrugs, etc.). Other types of molecules include, but are not limited to, small interfering ribonucleic acids, micro-RNAs, antisense oligonucleotides, aptamers, peptides, proteins (e.g., antibodies), naturally occurring organic or inorganic molecules, chemical elements (e.g., lithium), and any synthetic compounds greater or smaller than small molecules.
  • Component 2 may be incorporated into single dosage forms with Component 1 for combination therapy, which may be delivered by any safe, acceptable and effective route of administration, to block reconsolidation of dysfunctional neural circuit after Component 1 reactivates it to a labile state from its stable state.
  • Component 2 is selected from selective or non-selective receptor antagonists or inverse-agonists of the 5-HT2 receptors, selective or non-selective receptor antagonists or inverse-agonists of the 5-HT3 receptor, or selective or non-selective receptor antagonists or inverse-agonists of the NK-1 receptor, inclusive of all 5-HT2, 5-HT3 and NK-1 receptor subtypes, single nucleotide polymorphisms and other transcriptional, translational and post-transcriptional modifications of respective receptors expressed in humans.
  • ketanserin and its chemical homologues e.g., mianserin
  • mirtazapine and its chemical homologue e.g., setiptiline
  • ondansetron, granisetron, dolasetron, palonosetron and tropisetron e.g., ketanserin and its chemical homologues
  • mirtazapine and its chemical homologue e.g., setiptiline
  • ondansetron granisetron
  • dolasetron e.g., palonosetron and tropisetron.
  • any stereoisomers may be used alone or in combination in optimized proportions.
  • Component 2 may be also the substances described in Section 3 or modifications thereof, which are or are expected to be functional antagonists at the 5-HT3, 5-HT2 and/or NK-1 receptors based on: (1) their physiochemical characteristics, as evaluated by any analysis methods known to those skilled in the art, including but not limited to pharmacophore-based “docking”, virtual computational screening and other in vitro/in silico analysis methods; and/or (2) their in vitro and/or in vivo pharmacological profiles.
  • a 5-HT2 antagonist or an inverse-agonist is selected as component 2
  • an antagonist or an inverse-agonist selective for the 5-HT2A subtype, 5-HT2C subtype or both of the 5-HT2A and 5-HT2C subtypes is preferable to other antagonists or inverse-agonists selective for other 5-HT2 subtypes.
  • a 5-HT3 antagonist or an inverse-agonist may or may not show biochemical, in vitro and in vivo pharmacological properties or profiles, which are predictive of the substance being an antagonist, an inverse-agonist or a full or partial agonist at the ⁇ 7 nicotinic receptor.
  • a selective 5-HT3 antagonist is preferred over a 5-HT3 antagonists that is or is predicted to be a pharmacologically-effective ⁇ 7 nicotinic antagonist, an inverse agonist or a full or partial agonist.
  • NK-1 antagonist or inverse-agonist may or may not be selective for the NK-1 receptor subtype compared to the NK-2 and/or NK-3 receptor subtypes.
  • a non-selective NK-1 antagonist is preferable to a selective NK-1 antagonist.
  • Antagonists and pharmaceutically-acceptable salts and any modified forms thereof which exhibit a delayed and pulsatile release profile in any in vitro or in vivo tests that are well-known to those skilled in the art (e.g., in vitro dissolution test, direct determination of pharmacokinetic parameters in humans), are preferred over those with any other combinations/permutations of immediate, delayed, pulsatile, extended and sustained release profiles.
  • One preferred in vitro release profile of an antagonist is a dissolution rate of less than or equal to 10% in greater than or equal to 3 hours and a rate greater than or equal to 80% in less than or equal to 7 hours in simulated gastric or intestinal fluid.
  • Antagonists and pharmaceutically acceptable salts and modifications thereof may have a terminal elimination half-life (t1 ⁇ 2) of 20 hours or less in the human body.
  • a preferred terminal elimination half-life (t1 ⁇ 2) is 16 hours or less, and a more preferred terminal elimination half life (t1 ⁇ 2) is 14 hours or less. Even more preferred terminal elimination half life (t1 ⁇ 2) is 3 to 13 hours.
  • the maximum concentration of any drug following an in vivo administration is termed “Cmax,” and the time at which the Cmax is reached is termed “Tmax.”
  • Cmax The maximum concentration of any drug following an in vivo administration
  • Tmax the time at which the Cmax is reached.
  • the difference between the Tmax of a pharmacologically active form of component 1 and the Tmax of a pharmacologically active form of component 2 plays an essential role in determining the efficacy of combination therapy.
  • the Tmax separation between the plasma or serum concentration of a pharmacologically active form of component 1 and that of a pharmacologically active form of component 2 in humans may range from 1 hour to 12 hours.
  • a preferred Tmax separation is 1 to 8 hours, more preferably 2 to 8 hours, and even more preferably 2 to 7 hours.
  • Tmax of Component 1 and/or Component 2 may be modified through various methods including but not limited to: (1) the use of a pharmacologically-inactive prodrug that undergoes activation in, including but not limited to, the gastrointestinal track, blood, liver or other organs; or (2) formulation-based modification of the pharmacologically-effective parent form.
  • Various formulation-based methods including but not limited to pH-based, osmotic-based, liposome-based modification methods may be used to achieve the intended in vivo PK profiles in humans.
  • compositions for combination therapy may be provided as any dosage forms, including, but not limited to, tables, capsules, gels, suspensions, films, patches and suppositories.
  • the dosage forms may be pharmaceutical compositions containing component 1 and component 2 as active ingredients and may further contain pharmaceutically acceptable additives for formulation, such as excipients, carriers or the like.
  • the additives may be any pharmaceutically acceptable additives that may assist in formulation such as molding and facilitate intake.
  • the compositions may contain a predetermined amount of the active ingredients in powder or granules, a solution or a suspension in an aqueous or non-aqueous liquid, an oil-in-water or water-in-oil emulsion.
  • Such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and a suitable carrier which contains one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture.
  • a tablet may be prepared by compressing or molding a powder or granules containing the active compound, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form, such as a powder or granules optionally mixed with an excipient, such as a binder, lubricant, inert diluent, and/or surface active agent. Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
  • the present invention further encompasses anhydrous pharmaceutical compositions and dosage forms containing the active ingredient, since water can facilitate the degradation of some compounds.
  • water can facilitate the degradation of some compounds.
  • the addition of water is widely accepted in the pharmaceutical field as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time.
  • Anhydrous pharmaceutical compositions and dosage forms of the present invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms of the present invention, which contain lactose are preferably anhydrous if substantial contact with moisture and/or humidity-during manufacturing, packaging, and/or storage is expected.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained.
  • anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits.
  • suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
  • the active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier can take a wide variety of forms depending on the form of preparation desired for administration.
  • any of the usual pharmaceutical media can be employed as carriers, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, preferably without employing the use of lactose.
  • suitable carriers include powders, capsules, and tablets, with the solid oral preparations being preferred. If desired, tablets can be coated by standard aqueous or non-aqueous techniques.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
  • natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrol
  • suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • talc calcium carbonate
  • microcrystalline cellulose e.g., powdere., powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Disintegrants are used in the compositions of the present invention to provide tablets that disintegrate when exposed to an aqueous environment. An excessively large amount of a disintegrant will produce tablets which may disintegrate in the bottle. An excessively small amount of a disintegrant may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compound disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and is readily discernible to those of ordinary skill in the art. Typically, about 0.5 to about 15 wt % of disintegrant, preferably about 1 to about 5 wt % of disintegrant, may be used in the pharmaceutical composition.
  • Disintegrants that can be used to form pharmaceutical compositions and dosage forms include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, or mixtures thereof.
  • Lubricants which can be used to form the pharmaceutical compositions and dosage forms of the present invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof.
  • a lubricant may optionally be added in an amount of less than about 1 wt % of the pharmaceutical composition.
  • the essential active ingredient may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin, and various combinations thereof.
  • Tablets may be uncoated or coated by known techniques to delay disintegration and adsorption in the gastrointestinal tract to thereby provide a sustained reaction over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredients are mixed with an inert solid diluent, for example, lactose, mannitol, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredients are present as such, or mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil. Tablets may be disintegrated for quick release of the active ingredients.
  • Surfactants that may be used to form the pharmaceutical compositions and dosage forms of the present invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. Specifically, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
  • a suitable hydrophilic surfactant will generally have an HLB value of at least 10, while suitable lipophilic surfactants will generally have an HLB value of or less than about 10.
  • An empirical parameter commonly used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance (“HLB” value).
  • HLB hydrophilic-lipophilic balance
  • Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
  • Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.
  • lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10.
  • HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
  • Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixture
  • examples of ionic surfactants include lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; camitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • More preferred ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG-phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate,
  • Hydrophilic non-ionic surfactants include, but are not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene sterols, derivative
  • hydrophilic non-ionic surfactants include, but are not limited to, PEG-10 laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG-15 stearate, PEG-32 distearate, PEG-40 stearate, PEG-100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl ole
  • Suitable lipophilic surfactants include: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/vitamin derivatives; and mixtures thereof.
  • preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
  • the composition of the present invention may contain a solubilizer to ensure good solubilization or dissolution of the active ingredients, and to minimize precipitation of the active ingredients.
  • This solubilizer may be particularly important in compositions for parenteral use, for example, injectable compositions.
  • a solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other agents, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
  • solubilizers include, but are not limited to, alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether or methoxy PEG; amides and other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone, ⁇ -caprolactam, N-alkylpyrrolidone
  • solubilizers include, but are notlimited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide.
  • Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
  • the amount of solubilizer that can be included is not particularly limited.
  • the amount of a given solubilizer is limited to a biologically acceptable amount which may be readily determined by those skilled in the art.
  • the solubilizer can be in a weight ratio of 10 wt %, 25 wt %, 50 wt %, 100 wt %, or up to about 200 wt %, based on the combined weight of the active ingredients and other excipients.
  • solubilizer may also be used, such as 5%, 2%, 1% or less.
  • the solubilizer will be present in an amount of about 1 wt % to about 100 wt %, more typically about 5 wt % to about 25 wt %.
  • composition of the present invention may further contain one or more pharmaceutically acceptable additives and excipients.
  • additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavoring agents, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
  • an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons.
  • pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like.
  • bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like.
  • a pharmaceutically acceptable acid such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids
  • Salts of polyprotic acids such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate may also be used.
  • the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like. Preferred cations include sodium, potassium, lithium, magnesium, calcium and ammonium.
  • Suitable acids are pharmaceutically acceptable organic or inorganic acids.
  • suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like.
  • suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic
  • the present invention provides an injectable pharmaceutical composition containing an agent for reducing or eliminating the side effects of component 1 and component 2 as active ingredient, particularly component 1.
  • the present invention provides an injectable pharmaceutical composition containing a combination of an agent for reducing or eliminating the side effects of component 1 and an injectable pharmaceutical excipient. The content of the agent in the composition is as described herein.
  • Forms that may be used to administer the novel composition of the present invention include aqueous or oily suspensions or emulsions, sesame oil, corn oil, cottonseed oil, peanut oil, elixirs, mannitol, dextrose, sterile aqueous solutions and similar pharmaceutical media.
  • Aqueous solutions in saline are also conventionally used for injection.
  • Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed.
  • Proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like.
  • Sterile injectable solutions are prepared by incorporating the active ingredients in a required amount in an appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating various sterile active ingredients into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredients plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Sterile injectable solutions are prepared by incorporating the required amount of antibiotic(s) in the appropriate solvent with other various ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • compositions of the present invention can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical administration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, or dimethylsulfoxide (DMSO)-based solutions.
  • DMSO dimethylsulfoxide
  • high-density carriers are capable of providing an area exposed to the active ingredients over a long period of time.
  • a solution formulation may provide more immediate exposure of the active ingredients to the chosen area.
  • compositions may also contain suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
  • suitable solid or gel phase carriers or excipients which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
  • penetration-enhancing molecules known to those trained in the art of topical formulation.
  • humectants e.g., urea
  • glycols e.g., propylene glycol
  • alcohols e.g., ethanol
  • fatty acids e.g., oleic acid
  • surfactants e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.glycerol monolaurate, sulfoxides, terpenes (e.g., menthol)
  • amines amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • transdermal delivery devices e.g., patches or minipumps.
  • transdermal devices may be used to provide continuous or discontinuous infusion of the active ingredients in controlled amounts.
  • the invention provides a transdermal device incorporating an agonist and/or an antagonist.
  • transdermal devices for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such devices may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents, or mixtures thereof, and pharmaceutically acceptable powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described above.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • compositions in pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.
  • compositions described herein may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art.
  • Dosage forms for combination therapy may be administered by any routes and methods, including, but not limited to, intravenous, intranasal, inhalation (e.g., intrapulmonary), sublingual, transdermal, subcutaneous and intracranial routes.
  • routes and methods including, but not limited to, intravenous, intranasal, inhalation (e.g., intrapulmonary), sublingual, transdermal, subcutaneous and intracranial routes.
  • Neuropsychiatric disorders that may be treated with the combination therapy include PUD, LID and PTSD.
  • indications include but not limited to: (1) Other substance-related disorders, including but not limited to tobacco use disorder, alcohol use disorder and cannabis use disorder; (2) behavioral addictive disorders, including but not limited to compulsive gambling and internet addiction; (3) other trauma and stressor-related disorders, including but not limited to reactive attachment disorder, acute stress disorder, and adjustment disorder; (4) anxiety disorders, including but not limited to generalized anxiety disorder, panic disorder, agoraphobia, substance/medication-induced anxiety disorder; (5) obsessive-compulsive disorders, including but not limited to obsessive-compulsive disorder, body dysmorphic disorder, hoarding disorder, trichotillomania, and excoriation disorder; and (6) feeding and eating disorders, including but not limited to anorexia nervosa, bulimia nervosa and binge-eating disorder.
  • a combination dosage form may be used together with other therapies or dosage forms to achieve additional or synergistic effects compared to the combination dosage form given alone.
  • therapy or dosage forms that may be used combination therapy include, but not limited to behavioral therapies such as cognitive behavioral therapy, extinction therapy and exposure therapy, and drug therapies that use anti-epileptics, antipsychotics, antidepressants or ketamine.
  • the combination therapy may be used as a synergistic, additive, ameliorative and/or preventive adjunct treatment.
  • component 1 is methylphenidate or any immediate-release formulation of methylphenidate
  • component 2 is any delayed, pulsatile-release formulation of ondansetron.
  • Any combination/permutation of the doses of methylphenidate and ondansetron may be used.
  • the dose of methylphenidate in the dosage form is equivalent to 0.1 to 80 mg of methylphenidate hydrochloride
  • the dose of ondansetron in the dosage form is equivalent to 0.1 to 32 mg of ondansetron hydrochloride.
  • the dose of methylphenidate in the dosage form is equivalent to 1 to 80 mg of methylphenidate hydrochloride, and the dose of ondansetron in the dosage form is equivalent to 0.1 to 16 mg of ondansetron hydrochloride. Even more preferably, the dose of methylphenidate in the dosage form is equivalent to 5 to 60 mg of methylphenidate hydrochloride, and the dose of ondansetron in the dosage form is equivalent to 4 to 16 mg of ondansetron hydrochloride. Any permutation of the fixed doses of methylphenidate and ondansetron within the preferred dose ranges may be embodied in single dosage forms.
  • any stereoisomers may be used alone or in a combination thereof in optimized proportions.
  • the use of esmirtazapine, the S(+) stereoisomer of the antidepressant mirtazapine (e.g., Remeron®), as the Component 2 in dosage forms for treatment of LID is an example of preferred use of one stereoisomer over the others.
  • Mirtazapine is a tetracyclic, piperazino-azepine compound (1,2,3,4,10,14b-hexahydro-2-methylpyrazino[2,1-a]pyrido[2,3-c]benzazepine; C 17 H 19 N 3 ; molecular weight: 265.36), and is a noradrenergic and specific serotonergic antidepressant.
  • Commercially-available mirtazapine preparations e.g., Remeron®
  • Mirtazapine has high affinities to 5-HT2A and 5-HT2C receptors, as well as for 5-HT3, adrenergic ⁇ 2 and histamine H1 receptors. Furthermore, although this drug has minimal affinity to 5-HT1A receptors, the drug may also potentiate 5-HT1A-dependent neurotransmission through the blockade of ⁇ 2 adrenergic auto- and heteroreceptors and subsequent indirect increases in serotonin release. Since the drug is a 5-HT2 and 5-HT3 receptor antagonist, 5-HT1A-mediated serotonergic transmission is preferentially enhanced (functional 5-HT1A agonist).
  • esmirtazapine (1) shows higher affinity for 5-HT2A and 5-HT2C (and ⁇ 2 adrenergic) receptors; (2) may be absorbed faster (but not more extensively) by an “enantioselective” transport process in the gut wall; and (3) may accumulated more in the brain.
  • esmirtazapine is expected to be associated with lower incidences of untoward side effects such as excessive daytime somnolence compared to racemic mirtazapine (e.g., Remeron®) or the R( ⁇ ) enantiomer.
  • Esmirtazapine is predominantly metabolized by CYP2D6, which exhibits clinically-significant genetic polymorphism. Thus, its clearance is twice as low in poor metabolizers as compared to extensive metabolizers. Therefore, while any doses of esmirtazapine may be embodied in single combination therapy dosage forms, determination of the CYP2D6 phenotype in individual patients prior to the initiation of combination therapy may allow for a selection of pharmacogenetically-preferred esmirtazapine dose for individual patients (“individualized medicine”).
  • Ond-PR2 is a bead formulation prepared from ondansetron as the active pharmaceutical ingredient (API).
  • MPh-IR is an immediate-release generic formulation of methylphenidate.
  • [MPh-IR+Ond-PR2] is a single encapsulated dosage form consisting of MPh-IR and Ond-PR2 as Components 1 and 2, respectively.
  • Single capsules containing either [MPh-IR+Ond-PR2] or dextrose (placebo) were administered once a day for 14 days.
  • Subjects also underwent single fMRI-based assessments 1-7 days before the first study dosing (pre-treatment) and again between Day 3 and 7 days after the last dosing (i.e., post-treatment assessment was performed after complete drug washout).
  • Outcome measures for group comparisons were behavioral rating scales and blood-oxygenation-level-dependent (BOLD) functional imaging results under well-established resting-state, cue-reactivity and Go/noGO task paradigms. Twenty-eight subject completed the 2-week drug treatment and pre- and post-treatment fMRI assessments.
  • PK samples were collected following single oral administrations of a [MPh-IR+Ond-PR2] capsule to: (1) estimate the PK parameters of the two Components (i.e., MPh-IR and Ond-PR2] by quantifying serum methylphenidate and ondansetron concentrations; and (2) determine whether or not any of these parameters change after single daily administrations for 14 days.
  • Descriptive statistics for blood methylphenidate (MPh-IR) and ondansetron (Ond-PR2) concentrations at each time point were summarized as the mean, standard deviation and coefficient of variation. Estimation and statistical analyses of PK parameters were performed with SAS 9.3, and BABE Solution.
  • Table 1 presents the PK parameters of methylphenidate and ondansetron following a single oral administration of [MPh-IR+Ond-PR2] combination capsule on the first and last days of treatment (Days 1 and 14, respectively). No significant changes were observed in Cmax, Cmin, Tmax, t1 ⁇ 2, AUC (0-24), AUC (0-inf.), and Kel parameters for either methylphenidate or ondansetron between Day 1 and Day 14.
  • means the probability of obtaining the estimated value of mean group difference (e.g., ⁇ 1.63 for psychostimulant cue reactivity), when actual difference is 0.
  • mean group difference e.g., ⁇ 1.63 for psychostimulant cue reactivity
  • the probability for the combination therapy group and the placebo group to have the same mean score is only 3.3%.
  • 95% CI (95% Confidence Interval) means that there is a 95% confidence level that the true population mean difference between the combination therapy and the placebo groups falls between the low and high values (for example, ⁇ 3.12 to ⁇ 0.14 for compulsive cocaine scale). Confidence intervals which do not contain zero are declared significant difference between treatments with 95% confidence.
  • SJWS Shiffman-Jarvik Withdrawal Scale
  • the dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) was injected into the right medial forebrain bundle of rats to induce unilateral lesions of the nigrostriatal DA pathway.
  • 6-OHDA lesion animals were treated twice daily with levodopa methyl ester (25 mg/kg, i.p., bid immediately preceded by 6.5 mg/kg, i.p. benserazide) for 3 weeks to establish LIAIM.
  • Three hour LIAIM scores were quantified twice a week during the levodopa treatment using standard LIAIM rating scales.
  • the present invention may be used in the pharmaceutical industry and related industries, such as preparation of combination therapy compositions for treatment of neuropsychiatric disorders.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Addiction (AREA)
  • Psychiatry (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Dermatology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Nutrition Science (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Psychology (AREA)
  • Obesity (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Emergency Medicine (AREA)
  • Dispersion Chemistry (AREA)
  • Medicinal Preparation (AREA)
US15/662,289 2015-01-28 2017-07-28 Composition and method for treatment of neuropsychiatric disorders Abandoned US20170326127A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/662,289 US20170326127A1 (en) 2015-01-28 2017-07-28 Composition and method for treatment of neuropsychiatric disorders
US16/808,458 US20200197383A1 (en) 2015-01-28 2020-03-04 Composition and method for treatment of neurophychiatric disorders
US18/121,626 US20230293443A1 (en) 2015-01-28 2023-03-15 Composition and method for treatment of neuropsychiatric disorders

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201562108616P 2015-01-28 2015-01-28
PCT/KR2016/000903 WO2016122218A2 (ko) 2015-01-28 2016-01-28 신경정신계 장애의 치료를 위한 조성물 및 방법
US15/662,289 US20170326127A1 (en) 2015-01-28 2017-07-28 Composition and method for treatment of neuropsychiatric disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2016/000903 Continuation WO2016122218A2 (ko) 2015-01-28 2016-01-28 신경정신계 장애의 치료를 위한 조성물 및 방법

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/808,458 Continuation US20200197383A1 (en) 2015-01-28 2020-03-04 Composition and method for treatment of neurophychiatric disorders

Publications (1)

Publication Number Publication Date
US20170326127A1 true US20170326127A1 (en) 2017-11-16

Family

ID=56544502

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/662,289 Abandoned US20170326127A1 (en) 2015-01-28 2017-07-28 Composition and method for treatment of neuropsychiatric disorders
US16/808,458 Abandoned US20200197383A1 (en) 2015-01-28 2020-03-04 Composition and method for treatment of neurophychiatric disorders
US18/121,626 Pending US20230293443A1 (en) 2015-01-28 2023-03-15 Composition and method for treatment of neuropsychiatric disorders

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/808,458 Abandoned US20200197383A1 (en) 2015-01-28 2020-03-04 Composition and method for treatment of neurophychiatric disorders
US18/121,626 Pending US20230293443A1 (en) 2015-01-28 2023-03-15 Composition and method for treatment of neuropsychiatric disorders

Country Status (7)

Country Link
US (3) US20170326127A1 (ko)
EP (1) EP3251699B1 (ko)
JP (4) JP6655090B2 (ko)
KR (6) KR20190077590A (ko)
CN (2) CN107206093A (ko)
DK (1) DK3251699T3 (ko)
WO (1) WO2016122218A2 (ko)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102176538B1 (ko) 2019-01-31 2020-11-09 한국과학기술연구원 항정신병성 약물 검출용 fret 바이오센서 및 이를 이용한 검출 방법

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070134077A1 (en) * 2005-12-08 2007-06-14 Conestoga Cold Storage Rack, conveyor and shuttle automated pick system
US8512751B2 (en) * 2004-12-20 2013-08-20 Collegium Pharmaceutical, Inc. Pharmaceutical compositions for sleep disorders

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997003671A1 (en) * 1995-07-14 1997-02-06 Medeva Europe Limited Composition comprising d-threo-methylphenidate and another drug
WO2003037313A2 (en) * 2001-10-31 2003-05-08 Recovery Pharmaceuticals, Inc. Methods for the treatment of addiction
DE602004016288D1 (de) 2003-01-16 2008-10-16 Acadia Pharm Inc Selektive serotonin 2a/2c inverse rezeptoragonisten als therapeutika für neurodegenerative erkrankungen
AU2007249435A1 (en) * 2006-05-09 2007-11-22 Braincells, Inc. 5 HT receptor mediated neurogenesis
KR20110022631A (ko) * 2008-05-27 2011-03-07 인트라-셀룰라 써래피스, 인코퍼레이티드. 수면 장애 및 다른 장애를 치료하기 위한 방법 및 조성물
WO2010015029A1 (en) * 2008-08-06 2010-02-11 Gosforth Centre (Holdings) Pty Ltd Compositions and methods for treating psychiatric disorders
US20130116215A1 (en) * 2011-10-28 2013-05-09 Mireia Coma Combination therapies for treating neurological disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8512751B2 (en) * 2004-12-20 2013-08-20 Collegium Pharmaceutical, Inc. Pharmaceutical compositions for sleep disorders
US20070134077A1 (en) * 2005-12-08 2007-06-14 Conestoga Cold Storage Rack, conveyor and shuttle automated pick system

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Clarke and Speller, Cochrane Database of Systematic Reviews, Issue 2 (Year: 1999) *
Clinical Trial NCT01377662, published online at on June 21, 2011 Retrieved from: https://clinicaltrials.gov/ct2/show/NCT01377662 Retrieved on: 4/30/2019 (Year: 2011) *
Vanover et al., Pharmacol,Biochem, and Behav, 90:540-544 (Year: 2008) *

Also Published As

Publication number Publication date
WO2016122218A3 (ko) 2016-10-13
EP3251699A2 (en) 2017-12-06
KR20210095734A (ko) 2021-08-02
JP2021193118A (ja) 2021-12-23
JP7288016B2 (ja) 2023-06-06
KR20220071297A (ko) 2022-05-31
EP3251699A4 (en) 2018-08-01
KR20180015302A (ko) 2018-02-12
US20230293443A1 (en) 2023-09-21
JP2023109947A (ja) 2023-08-08
US20200197383A1 (en) 2020-06-25
DK3251699T3 (da) 2024-05-21
KR101994596B1 (ko) 2019-06-28
CN114146088A (zh) 2022-03-08
KR20160103548A (ko) 2016-09-01
WO2016122218A2 (ko) 2016-08-04
EP3251699B1 (en) 2024-04-17
JP6943994B2 (ja) 2021-10-06
KR20200032759A (ko) 2020-03-26
JP2018503677A (ja) 2018-02-08
JP2020075938A (ja) 2020-05-21
KR20190077590A (ko) 2019-07-03
CN107206093A (zh) 2017-09-26
JP6655090B2 (ja) 2020-02-26

Similar Documents

Publication Publication Date Title
EP1539149B1 (en) Combinations of metabotropic glutamate receptor antagonists and their use in treating addictive disorders
US20070087977A1 (en) Methods and compositions for treating pain
US20060111308A1 (en) Methods and compositions for therapeutic treatment
CN106456629A (zh) 用于运动机能亢进性运动障碍的治疗的vmat2抑制剂
GB2448224A (en) Combination of a nicotine receptor agonist and a dopaminergic agent with reduced side effects in the treatment of Parkinson's disease and the like
Summey Jr et al. Pharmacologic advances in the systemic treatment of itch
EA026869B1 (ru) Способ лечения идиопатического легочного фиброза
WO2016073294A1 (en) Method for treating underactive bladder syndrome
US20230293443A1 (en) Composition and method for treatment of neuropsychiatric disorders
WO2019104291A1 (en) Combination of granulated dried botanical extract powder for symptom relief
US9233109B2 (en) Compositions, methods of use, and methods of treatment
AU2016302769A1 (en) Antitussive compositions and methods
CN111479592A (zh) 阿片样物质使用障碍、阿片样物质戒断症状和慢性疼痛的治疗
Dassanayake et al. Replacement treatment during extinction training with the atypical dopamine uptake inhibitor, JHW-007, reduces relapse to methamphetamine seeking
MX2013004924A (es) Combinacion de bevacizumab y 2,2-dimetil-n((s)-6-oxo-6,7-dihidro-5 h-dibenzo[b,d] azepin-7-il)-n'-(2,2,3,3,3-pentafluoro-propil)-malo namida para el tratamiento de desordenes proliferativos.
ZA200507213B (en) Combination of desoxypeganine and mecamylamine forthe treatment of alcohol abuse
Merel Boom et al. NON ANALGESIC EFFECTS OF OPIOIDS: OPIOID-INDUCED RESPIRATORY DEPRESSION
GB2468424A (en) Composition for the reduction of side-effects of dopaminergic agents

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENERYS BIOPHARMACEUTICALS CORPORATION, NORTH CARO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, TONG HYON;REEL/FRAME:043122/0657

Effective date: 20170726

Owner name: GENERYS KOREA CORPORATION, KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, TONG HYON;REEL/FRAME:043122/0657

Effective date: 20170726

Owner name: DUKE UNIVERSITY, NORTH CAROLINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, TONG HYON;REEL/FRAME:043122/0657

Effective date: 20170726

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

AS Assignment

Owner name: DUKE UNIVERSITY, NORTH CAROLINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GENERYS KOREA CORPORATION;GENERYS BIOPHARMACEUTICALS CORPORATION;REEL/FRAME:049031/0961

Effective date: 20190125

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION