WO2020185712A1 - Methods for treating sleep fragmentation disorders - Google Patents

Methods for treating sleep fragmentation disorders Download PDF

Info

Publication number
WO2020185712A1
WO2020185712A1 PCT/US2020/021770 US2020021770W WO2020185712A1 WO 2020185712 A1 WO2020185712 A1 WO 2020185712A1 US 2020021770 W US2020021770 W US 2020021770W WO 2020185712 A1 WO2020185712 A1 WO 2020185712A1
Authority
WO
WIPO (PCT)
Prior art keywords
sleep
compound
disorder
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/US2020/021770
Other languages
French (fr)
Inventor
Dale M. Edgar
Original Assignee
Alairion, Inc.
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 Alairion, Inc. filed Critical Alairion, Inc.
Publication of WO2020185712A1 publication Critical patent/WO2020185712A1/en

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • 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
    • A61K31/4151,2-Diazoles
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/4468Non condensed piperidines, e.g. piperocaine having a nitrogen directly attached in position 4, e.g. clebopride, fentanyl
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the present disclosure relates to methods of alleviating a symptom of, treating, or preventing a sleep disorder (e.g., increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold) by administering a compound of the present disclosure, or pharmaceutically acceptable salt thereof.
  • a sleep disorder e.g., increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold
  • poor sleep Whilst the daytime impairment caused by poor sleep has long been appreciated, poor sleep also has cascading negative impact upon alertness, cognition, learning and memory, vigilance, performance, and a broad range of co-morbid health conditions including acute and chronic pam and pain disorders, psychiatric conditions, neurodegenerative disease, developmental disorders, metabolic disease and diabetes, obesity , cardiovascular disease, immunological disorders, and many other medical conditions.
  • the present disclosure provides compounds for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold. In some embodiments, the sleep disorder is increased disturbed sleep. In some embodiments, the sleep disorder is increased sleep fragmentation. In some embodiments, the sleep disorder is increased arousals. In some embodiments, the sleep disorder is decreased arousal threshold.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAs receptor agonist.
  • the compound is a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAs receptor
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a2d-1 and a2d-2 auxiliary subunits of voltage-gated calcium channels.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels. [0012] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a a2d-2 auxiliar subunit of voltage-gated calcium channels.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a 5HT:?A receptor antagonist and inverse agonist.
  • the present disclosure provides the identification of a pharmacological compound class, wlierein the compound is a 5HT2A receptor antagonist or a 5HT2A receptor inverse agonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a 5HT2A receptor antagonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a 5HT2A receptor inverse agonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor antagonist and inverse agonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor antagonist or a Hi receptor inverse agonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor antagonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor inverse agonist.
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a gamma-hydroxy butyrate (GHB) receptor ligand.
  • GLB gamma-hydroxy butyrate
  • the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a GABAB receptor agonist.
  • the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage gated calcium channels is selected from gabapentm, pregabalm, and mirogabalin.
  • the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage gated calcium channels is selected from gabapentm and pregabalin.
  • the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage gated calcium channels is gabapentin. In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage-gated calcium channels is pregabalin. In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage-gated calcium channels is mirogabalin.
  • the 5HT?.A receptor antagonists and inverse agonists is selected from trazodone, ne!otanserin, and pimavanserin.
  • the 5HT?.A receptor antagonist or 5HT:?A receptor inverse agonist is selected from trazodone, nelotanserin, and pimavanserin.
  • the Hi receptor antagonist and inverse agonist is selected from doxepm and diphenhydramine.
  • the Hi receptor antagonist or Hi receptor inverse agonist is selected from doxepin and diphenhydramine.
  • the Hi receptor antagonist and inverse agonist is doxepin.
  • the Hi receptor antagonist or Hi receptor inverse agonist is doxepin.
  • the gamma-hydroxy butyrate (GHB) receptor ligand is gamma- hydroxybutyrate.
  • the GABAB receptor agonist is gamma-hydroxybutyrate.
  • the gamma-hydroxybutyrate is the sodium or potassium salt.
  • a compound of the present disclosure is selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
  • a compound of the present disclosure is selected from gabapentin, pregabalin, trazodone, nelotanserin, pimavanserin, doxepin, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds of the present disclosure selected from gabapentin, pregabalin, mirogabalin, trazodone,
  • nelotanserin pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering gabapentin, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering pregabalin, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering mirogabalin, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering trazodone, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering nelotanserin, or a pharmaceutically acceptable salt thereof
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering pimavanserin, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering doxepin, or a
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering diphenhydramine, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respiratory disturbance index (RD1), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • RD1 respiratory disturbance index
  • PLMD periodic leg movement disorder
  • REM behavior disorder elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with restless legs syndrome.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a high respiratory disturbance index (RDI).
  • RDI respiratory disturbance index
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a neurological disease.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a circadian rhythm disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with pain.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with periodic leg movement disorder
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with REM behavior disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with elderly fragmented sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with age-related sleep fragmentation.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with post-menopausal sleep disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-rnorbid with substance abuse.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with substance abuse withdrawal.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with narcolepsy.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a mental disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with snoring.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with idiopathic hypersomnia.
  • the methods of the present disclosure the subject is administered a pharmaceutical composition.
  • the pharmaceutical composition comprises a compound of the present disclosure and an additional active agent.
  • the additional active agent is a sedative-hypnotic.
  • the sedative-hypnotic is selected from zolpidem, suvorexant, butabarbitai, quazepam, triazolam, tasimelteon, eszopiclone, temazepam, ramelteon,
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder by administering a pharmaceutical composition comprising a ligand of a2d-1 and a2d-2 auxiliary subunits of voltage-gated calcium channels, a 5HT2A receptor antagonists, a 5HT2A receptor inverse agonists, a Hi receptor antagonists, a Hi receptor inverse agonists, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or a pharmaceutically acceptable derivative thereof, in combination with an additional active agent.
  • a pharmaceutical composition comprising a ligand of a2d-1 and a2d-2 auxiliary subunits of voltage-gated calcium channels, a 5HT2A receptor antagonists, a 5HT2A receptor inverse agonists, a Hi receptor antagonists, a Hi receptor inverse agonists, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder by administering a pharmaceutical composition comprising a compound of the present disclosure, or a pharmaceutically acceptable derivative thereof, in combination with one or more additional active agent.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder by administering the compounds of the present disclosure and any additional active agents, if present, either horn somni, h.s. (at bedtime) or between about 0-4 hours before bedtime to a subject m need thereof.
  • compositions and methods of using such pharmaceutical therapies are provided herein for treating a sleep disorder.
  • Pharmaceutical therapies and methods of using such pharmaceutical therapies are provided for a sleep disorder in a subject with one or more co- morbid medical conditions.
  • these therapies and methods of using such therapies include pharmaceutical therapies to decrease disturbed sleep, decrease sleep fragmentation, decrease arousals, or increase arousal threshold.
  • a subject may have one or more of the following medical conditions: sleep apnea, restless legs syndrome, a high respiratory disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • RDI respiratory disturbance index
  • PLMD periodic leg movement disorder
  • REM behavior disorder elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • LMAi Locomotor Activity Intensity
  • LMAi Locomotor Activity Intensity
  • “uninterrupted bouts of sleep” (average sleep bout duration) refer to the average duration of all bouts of uninterrupted sleep that occurred each hour, measured in minutes.“Interruption” of sleep refers to 2 or more consecutive 10 second epochs of
  • wakefulness.“Interruption” of wake refers to 2 or more consecutive 10 second epochs of sleep. The value for the length of a bout that extends into the subsequent hour is assigned to the hour in which it begins. Analogous quantification is carried out for bouts of wakefulness. Sleep bout length may reflect the human tendency to awaken periodically through the night (such awakenings are normally not recalled), which in turn may be an important factor determining the restorative value of sleep in humans. Pre-eiinicai measures of sleep bout duration are also predictors of soporific efficacy in a subject (e.g., human).
  • NREM non-rapid eye movement sleep stages
  • “reduced number of arousals” refers to the reduced number of wake bouts per hour.
  • REM refers to the rapid eye movement sleep stage.
  • CT refers to circadian time
  • “sleep consolidation” refers to the measurement of the average sleep bout duration per hour.
  • “sleep continuity” refers to the measurement of sleep-bout length.
  • the“depth” of sleep is characterized by EEG slow wave activity, which may subserve sleep continuity or sleep consolidation, which is one of several determinants of sleep quality.
  • SWA slow wave activity
  • EEG delta po EEG delta po
  • LMA intensity refers to locomotor activity (LMA) counts per minute of EEG-defmed wakefulness. This variate allows an assessment of LMA that is independent of the amount of time awake, which may be used to quantify the specificity of a wake- or sleep-promoting effect.
  • number of wake bouts refers to the number of uninterrupted bouts of wakefulness that occurred each hour, measured in minutes. Number of wake bouts are of interest because it may closely reflects the number of arousals from sleep that occur at the time of measurement. Interpreted together, average sleep bout duration and number of wake bouts provide a highly reliable assessment of a drug or novel molecular entity (NME) effect on sleep fragmentation. Drugs that improve sleep fragmentation have been shown to improve the restorative benefits of sleep.
  • NME novel molecular entity
  • the term“co-morbid” refers to a disease or disorder which is concurrent with a sleep disorder, but may not be the cause, in whole or in part, of the sleep disorder.
  • the term“pharmaceutically acceptable” refers to compounds, anions, cations, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • compositions comprising any compound described herein in combination with at least one pharmaceutically acceptable excipient or carrier.
  • the term“pharmaceutical composition” is a formulation comprising a compound of the present disclosure in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial.
  • the quantity of active ingredient (e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved.
  • active ingredient e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof
  • the dosage will also depend on the route of administration.
  • routes of administration A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, mtrapentoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like.
  • Dosage forms for the topical or transdermal administration of a compound of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
  • the term“preventing,”“prevent,” or“protecting against” describes reducing or eliminating the onset of the symptoms or complications of such disease, condition or disorder.
  • references to“treating” or“treatment” include the alleviation of established symptoms of a condition.“Treating” or“treatment” of a state, disorder or condition therefore includes: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclimcal symptoms of the state, disorder or condition, (2) inhibiting the state, disorder or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof (in case of maintenance treatment) or at least one clinical or subciinical symptom thereof! or (3) relieving or attenuating the disease, i.e., causing regression of the state, disorder or condition or at least one of its clinical or subciinical symptoms.
  • the term“subject” is interchangeable with the term“subject m need thereof’, both of which refer to a subject having a disease or having an increased risk of developing the disease.
  • A“subject” includes a mammal.
  • the mammal can be , for example, a human or appropriate non-human mammal, such as primate, mouse, rat, dog, cat, cow, horse, goat, camel, sheep or a pig.
  • the subject can also be a bird or fowl.
  • the mammal is a rat.
  • the mammal is a human.
  • a subject in need has been previously diagnosed or identified as having a disease or disorder disclosed herein.
  • a subject in need thereof can also be one who is suffering from a disease or disorder disclosed herein.
  • a subject in need thereof can be one who has an increased risk of developing such disease or disorder relative to the population at large (i.e., a subject who is predisposed to developing such disorder relative to the population at large).
  • a subject in need thereof can have a refractory or resistant a disease or disorder disclosed herein (i.e., a disease or disorder disclosed herein that does not respond or has not yet responded to treatment). The subject may he resistant at start of treatment or may become resistant during treatment.
  • the subject in need thereof received and failed all known effective therapies for a disease or disorder disclosed herein.
  • the subject in need thereof received at least one prior therapy.
  • “approximately” and“about” are synonymous.
  • “approximately” and“about” refer to the recited value, amount, dose, or duration ⁇ 20%, ⁇ 15%, ⁇ 10%, ⁇ 8%, ⁇ 6%, ⁇ 5%, ⁇ 4%, ⁇ 2%, ⁇ 1%, or ⁇ 0.5%.
  • “approximately” and“about” refer to the listed value, amount, dose, or duration ⁇ 10%, ⁇ 8%, ⁇ 6%, ⁇ 5%, ⁇ 4%, or ⁇ 2%.
  • “approximately” and“about” refer to the listed value, amount, dose, or duration ⁇ 5%. In some embodiments,“approximately” and“about” refer to the listed value, amount, dose, or duration ⁇ 2%. In some embodiments,“approximately” and“about” refer to the listed amount, value, amount, dose, or duration ⁇ 1%.
  • a“compound” is the same as an“active ingredient”,“a ligand of «25-1 and «25-2 auxiliary' subunits of voltage-gated calcium channels, a 5HT2A receptor antagonists, a 5HT2A receptor inverse agonists, a Hi receptor antagonists, a Hi receptor inverse agonists, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAs receptor agonist”,“a ligand of «25-1 and «25-2 auxiliary' subunits of voltage-gated calcium channels”,“a 5HT2A receptor antagonists”,“a 5HT2A receptor inverse agonists”,“a Hi receptor antagonists”,“a Hi receptor inverse agonists”,“gamma-hydroxy butyrate (GHB) receptor ligand”,“a GABAs receptor agonist”, and a“compound of the present disclosure”.
  • “gabapentin” may also be referred to as HY-10310 or 2-[l-
  • “pregabaiin” may also be referred to as HY- 10204 or (3S)-3-
  • “mirogabalin” may also be referred to as 2-[(lR,5S,6S)-6-
  • “trazodone” may also be referred to as 2- ⁇ 3-[4-(3- chlorophenyl)piperazm- 1 -yl]propyl ⁇ -2H,3H- [ 1 ,2,4]triazolo[4,3 -a]pyndin-3 -one (i. e. ,
  • “nelotansenn” may also be referred to as 3-[3-(4-bromo-l-methyl- lH-pyrazol-5-yl)-4-methoxyphenyl]-l-(2,4-difluorophenyl)urea (i.e.,
  • “pimavanserm” may also be referred to as l-[(4- fluorophenyl)methyl]-l-(l -methylpipendin-4-yl)-3- ⁇ [4-(2-methylpropoxy)phenyl]methyl ⁇ urea
  • “doxepm” may also he referred to as 3-(6H-
  • “diphenhydramine” may also be referred to as 2-(benzhydryloxyj-
  • N,N-dimethylethan- 1 -amine pharmaceutically acceptable salt thereof.
  • “gamma-hydroxybutyrate” may also he referred to as 4-
  • hydroxybutanoic acid i.e., or a pharmaceutically acceptable salt thereof.
  • gamma- hydroxy butyrate is a pharmaceutically acceptable salt.
  • gamma-hydroxybutyrate is the sodium salt.
  • gamma-hydroxybutyrate is the potassium salt.
  • “zolpidem” may also be referred to as Ambien®, HY- 10131, N,N-dimethyl-2-[6-methyl-2-(4-methylphenyl)imidazo[l,2-a]pyridin-3-yl]acetamide (i.e.,
  • zolpidem is a pharmaceutically acceptable salt.
  • zolpidem is zolpidem tartrate.
  • “S-zopiclone” may also be referred to as Lunesta®, eszopiclone, [(7S)-6-(5-chioiOpyridin-2-yl)-5-oxo-7H-pyrroio[3,4-b]pyrazin-7-yTj 4-methylpiperazine-l -
  • “suvorexant” may also be referred to as Belsomra®, [(7R)-4-(5- chloro ⁇ l,3-benzoxazol-2-yl) ⁇ 7-methyl ⁇ l,4-diazepan-i-yl]-[5-methyl ⁇ 2-(tnazol-2-
  • Sleep fragmentation a condition in humans characterized by poor sleep consolidation, frequent brief arousals or microarousals (defined by the American Academy of Sleep Medicine as episodes of cortical EEG activation lasting at least 2 seconds and up to 16 seconds in duration and interrupting sleep), and frequent transitions to lighter stages of sleep, results in significant daytime impairment that may include impaired attention and concentration, excessive sleepiness, impaired j udgement, impaired memory and learning, increased risk of accidents, and secondary morbidity and mortality when sleep fragmentation is a concomitant of pain, sleep disordered breathing, and other disease states. Patients suffering from sleep fragmentation are often unaware of the hundreds of brief arousals that may occur during the night, and primarily complain of severe daytime impairment. Sleep fragmentation patients often complain that their sleep is not beneficial, refreshing, or restorative.
  • insomnia is a separate and distinct medical diagnosis.
  • Insomnia is typically characterized by patient awareness and dissatisfaction with their sleep.
  • Most insomnia patients have a hyperarousal disorder that makes it difficult to fall asleep and/or difficult to stay asleep but enable them to function well during the daytime.
  • Insomnia is diagnosed by measuring the latency to persistent sleep, i.e., LPS (LPS of >30 minutes satisfies the definition of sleep-onset insomnia) and/or measuring the amount of time awake after sleep onset, i.e., WASQ (WASO of >50 minutes satisfies the definition of sleep-maintenance insomnia).
  • LPS LPS of >30 minutes satisfies the definition of sleep-onset insomnia
  • WASQ WASO of >50 minutes satisfies the definition of sleep-maintenance insomnia
  • insomnia patients and particularly the elderly, to awaken in the middle of the night and be unable to return to sleep. Unlike patients suffering from sleep fragmentation, insomnia patients are almost always highly aware of their inability to fall asleep or stay asleep at night and complain about their nighttime experience, and typically do not complain about daytime impairment.
  • the present disclosure provides for the use of a preclinical sleep- wake and physiological assay system, named SCORE-2000® , or“SCORETM”.
  • SCORETM pharmacology database includes standardized sleep-wake, physiological, and behavioral data for over 500 distinct compounds.
  • the standardized nature of the experimental designs, data quality' control, and data analysis methods enable qualitative and quantitative comparisons between compounds.
  • the present disclosure pertains to the identification of pharmacological compounds that are useful for treating sleep disorders characterized in whole or m part by sleep fragmentation.
  • rat sleep and human sleep have all of the necessary fundamental similarities to permit the rat to be used as a preclinical model.
  • compounds that are soporific in a rat may have soporific effects in a human, and compounds that are soporific in a human may have soporific effects in a rat [0145] Both rat and human exhibit robust circadian modulation of sleep tendency and sleep architecture.
  • The“homeostatic” control of sleep shares similarity across mammalian species, including humans, in that loss of sleep increases a homeostatic drive for sleep evidenced by a reduction in latency to sleep onset, increase in the depth of sleep that can be reflected by the amount of low- frequency“delta” EEG (“EEG slow waves”) during nonREM, an increase in sleep consolidation as measured by sleep bout duration, or an increase in total sleep time.
  • Sleep deprivation m a subject causes the subject to fall asleep faster, sleep deeper, sleep more efficiently (e.g., more consolidated bouts of sleep), and sleep more (e.g., an increase of sleep time) until the homeostatic drive for sleep becomes adequately discharged through the sleeping process.
  • Uninterrupted, well consolidated sleep can determine sleep quality in both a rat and a human. No matter how much a subject sleeps or what frequency of EEG dominates during sleep, the beneficial work of the sleeping process requires that sleep is not fragmented (interrupted) by frequent arousa!s.
  • compounds of the present disclosure which affect REM sleep by decreasing the latency to sleep onset, increasing sleep time, increasing the depth and/or consolidation of sleep, reducing arousals, or a combination of the aforementioned effects in a subject may have the same effects in a different subject.
  • the compounds of the present disclosure, which affect REM sleep by decreasing the latency to sleep onset effects in a subject may have the same effects on a subject of a different species.
  • the compounds of the present disclosure, which affect REM sleep by decreasing the latency to sleep onset effects in rats may have the same effects on a subject of different species.
  • the compounds of the present disclosure, which affect REM sleep by decreasing the latency to sleep onset effects in rats may have the same effects on a human.
  • compounds of the present disclosure which affect REM sleep by increasing sleep time m a subject have the same effects in a different subject.
  • the compounds of the present disclosure winch affect REM sleep by increasing sleep tune m a subject may have the same effects on a subject of a different species.
  • the compounds of the present disclosure which affect REM sleep by increasing sleep time in rats may have the same effects on a subject of a different species.
  • the compounds of the present disclosure which affect REM sleep by increasing sleep time in rats may have the same effects on a human.
  • compounds of the present disclosure winch affect REM sleep by increasing the depth and/or consolidation of sleep m a subject, have the same effects in a different subject.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth and/or consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth and/or consolidation of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth and/or consolidation of sleep in rats may have the same effects m a human.
  • compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in rats may have the same effects in a human.
  • compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in rats may have the same effects in a human.
  • compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in rats may have the same effects in a human.
  • compounds of the present disclosure winch affect REM sleep by increasing the consolidation of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect REM sleep by increasing the consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the consolidation of sleep m rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by increasing the consolidation of sleep m rats may have the same effects in a human.
  • compounds of the present disclosure winch affect REM sleep by reducing arousals in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect REM sleep by reducing arousals in a subject have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by reducing arousals in rats have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect REM sleep by reducing arousals in rats have the same effects in a human.
  • compounds of the present disclosure which affect NREM sleep by decreasing the latency to sleep onset, increasing sleep time, increasing the depth and/or consolidation of sleep, reducing arousals, or a combination of the aforementioned effects in a subject may have the same effects in a different subject.
  • the compounds of the present disclosure, which affect NREM sleep by decreasing the latency to sleep onset effects in a subject may have the same effects on a subject of a different species.
  • the compounds of the present disclosure, which affect NREM sleep by decreasing the latency to sleep onset effects in rats may have the same effects on a subject of different species.
  • the compounds of the present disclosure, which affect NREM sleep by decreasing the latency to sleep onset effects in rats may have the same effects on a human.
  • compounds of the present disclosure which affect NREM sleep by increasing sleep time in a subject have the same effects in a different subject.
  • the compounds of the present disclosure which affect NREM sleep by increasing sleep time in a subject may have the same effects on a subject of a different species.
  • the compounds of the present disclosure which affect NREM sleep by increasing sleep time in rats may have the same effects on a subject of a different species.
  • the compounds of the present disclosure which affect NREM sleep by increasing sleep time in rats may have the same effects on a human.
  • compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep m a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep in rats may have the same effects in a human.
  • compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep m a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep in rats may have the same effects in a human
  • compounds of the present disclosure which affect NREM sleep by increasing the depth or consolidation of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect NREM sleep by- increasing the depth or consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth or consolidation of sleep in rats may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth or consolidation of sleep in rats may have the same effects in a human.
  • compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in rats may have the same effects m a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in rats may have the same effects in a human.
  • compounds of the present disclosure which affect NREM sleep by increasing the consolidation of sleep in a subject have the same effects in a different subject.
  • Compounds of the present disclosure winch affect NREM sleep by increasing the consolidation of sleep in a subject may have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by increasing the consolidation of sleep in rats may have the same effects m a subject of a different species.
  • Compounds of the present disclosure wiiich affect NREM sleep by increasing the consolidation of sleep in rats, may have the same effects in a human.
  • compounds of the present disclosure which affect NREM sleep by reducing arousals in a subject have the same effects in a different subject.
  • Compounds of the present disclosure which affect NREM sleep by reducing arousals in a subject have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by reducing arousals in rats have the same effects in a subject of a different species.
  • Compounds of the present disclosure which affect NREM sleep by reducing arousals in rats have the same effects in a human.
  • sleep continuity can be measured as the duration of NREM“bouts” or the duration of REM bouts, or the duration NREM+REM“bouts”, wherein an arousal or bout of wakefulness interrupts the NREM-REM cycle.
  • sleep bout can be comprised of NREM, REM, or NREM+REM.
  • sleep bout can be comprised of NREM. In some embodiments, sleep bout can be comprised of REM. In some embodiments, sleep bout can be comprised of
  • NREM and REM sleep alternate in what may be called the NREM-REM cycle. In some embodiments NREM precedes REM.
  • the proportion of time spent in NREM versus REM is the same for different subjects. In some embodiments, the proportion of time spent in NREM versus REM is the same for different subjects of different species. In some embodiments, the proportion of time spent in NREM versus REM is the same for a rat and a subject of a different species. In some embodiments, the proportion of time spent in NREM versus REM is the same for a rat and a human.
  • the proportion of time spent in NREM versus REM is about 5: 1.
  • the proportion of time spent m NREM versus REM is about 4: 1. In some embodiments, the proportion of time spent in NREM versus REM is about 3: 1. In some embodiments, the proportion of time spent in NREM versus REM is about 2: 1.
  • the proportion of time spent in NREM versus REM is from about 100: 1 to about 1 : 1. In some embodiments, the proportion of time spent m NREM versus REM is from about 90: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 80: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 70: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 60: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 50: 1 to about 1 : 1.
  • the proportion of time spent in NREM versus REM is from about 40: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 30: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 20: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 10: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 5: 1 to about 1 : 1. In some embodiments, the species is a mouse. In some embodiments, the species is a hoofed animal. In some embodiments, the hoofed animal is a horse or cow. In some embodiments, the species is not a laboratory rat. In some embodiments, the species is not a human.
  • hypnotics reduce REM sleep to some degree, and several classes of sleep disorder medicines can strongly suppress REM sleep.
  • REM sleep suppression is relevant to learning, memory, and/or psychiatric health.
  • the relative effect of some classes of medicines for sleep disorders, neuropsychiatric disorders, and cardiovascular disease that either inhibit or stimulate REM sleep translates from a subject to a different subject.
  • the relative effect of some classes of medicines for sleep disorders, neuropsychiatric disorders, and cardiovascular disease that either inhibit or stimulate REM sleep translates from a subject to a subject of a different species.
  • the relative effect of some classes of medicines for sleep disorders, neuropsychiatric disorders, and cardiovascular disease that either inhibit or stimulate REM sleep translates from a rat to a subject of a different species.
  • the relative effect of some classes of medicines for sleep disorders that either inhibit or stimulate REM sleep translate from rat to a human.
  • a class of medicine which may inhibit or stimulate REM or NREM sleep is selected from a sleep therapeutic, a neuropsychiatric antidepressant, and a cardiovascular medicine.
  • a sleep therapeutic is a sedative hypnotic GABAA positive allosteric modulators that binds to the GABAA benzodiazepine receptor.
  • a neuropsychiatric antidepressant is a selective serotonin reuptake inhibitor (SSRI) or an atypical antipsychotics.
  • SSRI selective serotonin reuptake inhibitor
  • a cardiovascular medicine is a therapeutic that may bind to an alpha-adrenergic receptor.
  • rat and human sleep There are two differences which may be present between rat and human sleep. First, rats are night-active, whereas humans are day-active. This difference may have no importance per se for testing drug effects on sleep and wakefulness. The timing of the dose relative to the normal sleep period can be relied upon when evaluating drug efficacy on sleep related variables (e.g., inhibition of REM sleep) when comparing rat and human sleep.
  • sleep related variables e.g., inhibition of REM sleep
  • rats The difference between rats and humans is sleep-bout length, also referred to as“sleep continuity”
  • humans may consolidate sleep into a single period per day, interrupted normally only by short (e.g., less than 2 hours, less than 1 hour, less than 45 minutes, less than 30 minutes, less than 25 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 5 minutes, or less than 1 minute) bouts of wakefulness.
  • the abnormal conditions may result in human sleep becoming highly fragmented, diminishing the restorative benefits of sleep.
  • Rats may have shorter bouts of sleep that occur throughout the 24-hour day (e.g., on average, every 20 minutes, a rat completes a sleep-wake cycle).
  • sleep typically occupies about 1/3 of each 20-minute cycle, and REM sleep is rare.
  • the rat typically sleeps about 2/3 of each 20-minute cycle.
  • the polyphasic nature of sleep and shorter spontaneous sleep bout durations m the rat, enables highly sensitive assessments of drug effects, such as those that increase sleep consolidation (sleep bout duration), decrease the number of arousals (number of wake bouts), and a variety of secondary but desirable measures of sleep quality, for example, EEG slow wave activity in noriREM sleep, and measures of wake maintenance as measured by wake bout duration.
  • Sleep bout-length may also be a sensitive measure of physiological sleepiness and is a pre-clinical predictor of soporific efficacy in humans.
  • Empirical optimization can be performed by assessing sleep-related compounds by administering such compounds at two circadian times of day, CT-18 and CT-5, wherein CT-0 is defined as lights-on.
  • CT-18 is the mid-point of the activity phase of the rat’s circadian cycle, 6 hours after lights-off, and may be especially sensitive to soporific drug effects on sleep bout length, although such effects can be observed at both CT-18 and CT-5.
  • CT-5 begins several hours of peak abundance of REM sleep and thus is a sensitive time to reveal drug-related inhibition of REM sleep.
  • Both CT-18 and CT-5 are suitable times of the day for the assessment of drug effects on sleep fragmentation as measured by arousals (number of wake bouts), sleep consolidation (sleep bout duration), as well as assessments of maintenance of wakefulness (wake bout length) and drug-related side effects.
  • the subject is surgically prepared for EEG and EMG recording and administered an analgesic with an antibiotic, followed by therapeutic deliver via intrapentoneal or oral administration.
  • the sleep and wakefulness is determined using SCORETM.
  • the sleep disorder is improved by (i) reducing the number of arousals fas measured by the number of wake bouts per hour), and (if) increasing sleep consolidation (as measured by average sleep bout duration per hour).
  • the sleep disorder is improved by (i) reducing the number of arousals (as measured by the number of wake bouts per hour), or (ii) increasing sleep consolidation (as measured by average sleep bout duration per hour).
  • the sleep disorder is improved by reducing the number of arousals (as measured by the number of wake bouts per hour.
  • sleep fragmentation is improved by increasing sleep consolidation (as measured by average sleep bout duration per hour).
  • the present disclosure is directed to a method of alleviating a symptom of treating, or preventing a sleep disorder.
  • the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold.
  • the sleep disorder is increased disturbed sleep. In some embodiments, the sleep disorder is increased sleep fragmentation. In some embodiments, the sleep disorder is increased arousals. In some embodiments, the sleep disorder is decreased arousal threshold.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds of the present disclosure or pharmaceutically acceptable salt thereof to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist, a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
  • a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels
  • a 5HT2A receptor antagonist a 5HT2A receptor inverse agonist
  • Hi receptor antagonist a Hi receptor inverse agonist
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels or a2d-2 auxiliary subunit of voltage-gated calcium channels, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 and a2d- 2 auxiliary subunits of voltage-gated calcium channels, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, or pharmaceutically acceptable salts thereo to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a 5HT2A receptor antagonist and inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a 5HT2A receptor antagonist or a 5HT2A receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a 5HT2A receptor antagonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a SHTZA receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor antagonist and inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor antagonist or a Hi receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor antagonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a a2d-2 auxiliary subunit of voltage-gated calcium channels, a gamma-hydroxy butyrate (GHB) receptor ligand, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • GLB gamma-hydroxy butyrate
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a GABAB receptor agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of or treating a sleep disorder by administering one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of a sleep disorder by administering one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxy butyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserm, and doxepin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
  • the present disclosure is directed to a method of alleviating a symptom of or treating a sleep disorder by administering one or more compounds selected from gabapentin, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a
  • the present disclosure is directed to a method of alleviating a symptom of a sleep disorder by administering one or more compounds selected from gabapentin, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure provides one or more compounds selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use m alleviating a symptom of or treating a sleep disorder.
  • the present disclosure provides one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use m alleviating a symptom of a sleep disorder.
  • the present disclosure provides one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of or treating a sleep disorder.
  • the present disclosure provides one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, mirogabalm, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of or treating a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of or treating a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gahapentin, pregabafm, mirogabalin, trazodone, nelotanserm, pimavanserm, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of or treating a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserm, and doxepin, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of or treating a sleep disorder.
  • the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserm, and doxepin, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of sleep a sleep disorder.
  • the compound is gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, or gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
  • the compound is gabapentm, pregabalin, or mirogabalin, or a pharmaceutically acceptable salt thereof.
  • the compound is gabapentm or pregabalm, or a pharmaceutically acceptable salt thereof.
  • the compound is trazodone, nelotanserm, or pimavanserin, or a pharmaceutically acceptable salt thereof.
  • the compound is doxepin or diphenhydramine, or a
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering gabapentin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
  • the present disclosure provides gabapentin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of gabapentin, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of gabapentin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is gabapentm, or a pharmaceutically acceptable salt thereof.
  • gabapentin is administered at a dose between about 0.010 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 0.05 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 0.1 mg/kg and about 500 mg/kg. In some embodiments, gabapentm is
  • gabapentm is administered at a dose between about 0.5 mg/kg and about 500 mg/kg. In some embodiments, gabapentm is administered at a dose between about 1 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 5 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 400 mg/kg. In some embodiments, gabapentm is administered at a dose between about 10 mg/kg and about 300 mg/kg.
  • gabapentin is administered at a dose between about 10 mg/kg and about 200 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 100 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 400 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 300 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 200 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 100 mg/kg. In some embodiments, gabapentin is
  • gabapentin is administered at a dose between about 75 mg/kg and about 300 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 200 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 150 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 125 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 100 mg/kg.
  • gabapentin is administered at a dose of about 0.010 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.015 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.020 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.025 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.05 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.1 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.2 mg/kg.
  • gabapentin is administered at a dose of about 0.3 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.4 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.5 mg/kg. In some embodiments, gabapentin is administered at a dose of about 1 mg/kg. In some embodiments, gabapentin is administered at a dose of about 2 mg/kg. In some embodiments, gabapentin is administered at a dose of about 3 mg/kg. In some embodiments, gabapentin is administered at a dose of about 4 mg/kg. In some embodiments, gabapentin is administered at a dose of about 5 mg/kg.
  • gabapentin is administered at a dose of about 6 mg/kg. In some embodiments, gabapentin is administered at a dose of about 7 mg/kg. In some embodiments, gabapentin is administered at a dose of about 8 mg/kg. In some embodiments, gabapentin is administered at a dose of about 9 mg/kg. In some embodiments, gabapentin is administered at a dose of about 10 mg/kg. In some embodiments, gabapentin is administered at a dose of about 15 mg/kg. In some embodiments, gabapentin is administered at a dose of about 20 mg/kg. In some embodiments, gabapentm is administered at a dose of about 25 mg/kg.
  • gabapentin is administered at a dose of about 50 mg/kg. In some embodiments, gabapentin is administered at a dose of about 75 mg/kg. In some embodiments, gabapentm is administered at a dose of about 100 mg/kg. In some embodiments, gabapentin is administered at a dose of about 125 mg/kg. In some embodiments, gabapentin is administered at a dose of about 150 mg/kg. In some embodiments, gabapentin is administered at a dose of about 175 mg/kg. In some embodiments, gabapentin is administered at a dose of about 200 mg/kg.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering pregabalin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
  • the present disclosure provides pregabalin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of pregabalin, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of pregabalin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is pregabalin, or a pharmaceutically acceptable salt thereof.
  • pregabalin is administered at a dose between about 0.010 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 0.05 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 0.1 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 0.5 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 45 mg/kg.
  • pregabalin is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 35 mg/kg. In some embodiments, pregabalin is administered at a dose between 1 mg/kg and 30 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 25 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 20 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 15 mg/kg.
  • pregabalin is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 25 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 20 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 15 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 30 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 25 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 20 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 15 mg/kg.
  • pregabalin is administered at a dose of about 0.010 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.015 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.020 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.025 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.05 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.1 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.2 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.3 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.4 mg/kg. In some
  • pregabalin is administered at a dose of about 0.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 1 mg/kg. In some embodiments, pregabalin is administered at a dose of about 1.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 2 mg/kg. In some embodiments, pregabalin is administered at a dose of about 2.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 3 mg/kg. In some embodiments, pregabalin is administered at a dose of about 3.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 4 mg/kg.
  • pregabalin is administered at a dose of about 4.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 10 mg/kg. In some embodiments, pregabalin is administered at a dose of about 15 mg/kg. In some embodiments, pregabalin is administered at a dose of about 20 mg/kg. In some embodiments, pregabalm is administered at a dose of about 25 mg/kg. In some embodiments, pregabalin is administered at a dose of about 30 mg/kg. In some embodiments, pregabalin is administered at a dose of about 35 mg/kg.
  • pregabalin is administered at a dose of about 40 mg/kg. In some embodiments, pregabalin is administered at a dose of about 45 mg/kg. In some embodiments, pregabalin is administered at a dose of about 50 mg/kg.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering mirogabalin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
  • the present disclosure provides mirogabalin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of mirogabalin, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of mirogabalin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is mirogabalin, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering trazodone, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure provides trazodone, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of trazodone, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of trazodone, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is trazodone, or a pharmaceutically acceptable salt thereof.
  • trazodone is administered at a dose between about 0.010 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 0.05 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 0.1 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 0.5 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 90 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 80 mg/kg.
  • trazodone is administered at a dose between about 1 mg/kg and about 70 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 60 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 30 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 100 mg/kg.
  • trazodone is administered at a dose between about 5 mg/kg and about 90 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 80 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 70 mg/kg.
  • trazodone is administered at a dose between about 5 mg/kg and about 60 rng/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 50 rng/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 60 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 50 mg/kg.
  • trazodone is administered at a dose between about 10 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 30 g/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 60 g/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 30 mg/kg.
  • trazodone is administered at a dose between about 20 mg/kg and about 60 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 30 mg/kg.
  • trazodone is administered at a dose of about 0.010 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.015 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.020 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.025 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.05 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.1 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.2 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.3 mg/kg. In some
  • trazodone is administered at a dose of about 0.4 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.5 mg/kg. In some embodiments, trazodone is administered at a dose of about 1 mg/kg. In some embodiments, trazodone is administered at a dose of about 2 mg/kg. In some embodiments, trazodone is administered at a dose of about 3 mg/kg. In some embodiments, trazodone is administered at a dose of about 4 mg/kg. In some embodiments, trazodone is administered at a dose of about 5 mg/kg. In some embodiments, trazodone is administered at a dose of about 10 mg/kg.
  • trazodone is administered at a dose of about 15 mg/kg. In some embodiments, trazodone is administered at a dose of about 20 mg/kg. In some embodiments, trazodone is administered at a dose of about 25 mg/kg. In some embodiments, trazodone is administered at a dose of about 30 mg/kg. In some embodiments, trazodone is administered at a dose of about 35 mg kg. In some embodiments, trazodone is administered at a dose of about 40 mg/kg. In some embodiments, trazodone is administered at a dose of about 45 mg/kg. In some embodiments, trazodone is administered at a dose of about 50 mg/kg.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering nelotanserin, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure provides nelotansenn, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of nefotanserin, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of nelotanserin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is nelotanserin, or a pharmaceutically acceptable salt thereof.
  • nelotanserin is administered at a dose between about 0.010 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 0.05 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 0.1 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is
  • nelotanserin is administered at a dose between about 0.5 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 45 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 35 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 30 mg/kg.
  • nelotanserin is administered at a dose between about 1 mg/kg and about 25 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 20 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 15 mg/kg. In some embodiments, nelotansenn is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 5 mg/kg and about 25 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 5 mg/kg and about 20 mg/kg.
  • nelotanserin is administered at a dose between about 5 mg/kg and about 15 mg/kg. In some embodiments, nelotansenn is administered at a dose between about 10 mg/kg and about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 10 nig/kg and about 25 nig/kg. In some embodiments, nelotanserin is administered at a dose between about 10 mg/kg and about 20 mg/kg. In some embodiments, nelotansenn is administered at a dose between about 10 mg/kg and about 15 mg/kg.
  • nelotanserin is administered at a dose of about 0.010 g kg. In some embodiments, nelotanserin is administered at a dose of about 0.015 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.020 mg/kg. In some
  • nelotanserin is administered at a dose of about 0.025 mg/kg.
  • nelotanserin is administered at a dose of about 0.05 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.1 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.2 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.3 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.4 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 1 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.05 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.1 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.2 mg/kg. In some embodiments, nelotans
  • nelotanserin is administered at a dose of about 1.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 2 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 2.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 3 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 3.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 4 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 4.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 5 mg/kg.
  • nelotanserin is administered at a dose of about 10 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 15 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 20 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 25 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 35 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 40 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 45 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 50 mg/kg.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering pimavanserin, or a pharmaceutically acceptable salt thereof, to a subject in need thereof [0264] In some embodiments, the present disclosure provides pimavanserm, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of pimavanserin, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of pimavanserin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is pimavanserin, or a pharmaceutically acceptable salt thereof.
  • pimavanserin is administered at a dose between about 0.010 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 0.05 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 0.1 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 0.5 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 400 mg/kg.
  • pimavanserin is administered at a dose between about 1 mg/kg and about 300 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 1 mg/kg and about 200 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 1 mg/kg and about 100 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 60 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 10 mg/kg and about 500 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 10 mg/kg and about 400 mg/kg.
  • pimavanserin is administered at a dose between about 10 mg/kg and about 300 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 10 mg/kg and about 200 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 10 mg/kg and about 100 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 10 mg/kg and about 60 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 30 mg/kg and about 100 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 30 mg/kg and about 60 mg/kg.
  • pimavanserin is administered at a dose of about 0.010 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.01 5 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.020 mg/kg. In some
  • pimavanserin is administered at a dose of about 0.025 mg/kg.
  • pimavanserin is administered at a dose of about 0.05 mg/kg.
  • pimavanserin is administered at a dose of about 0.1 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.2 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.3 mg/kg. In some embodiments, pimavanserin is
  • pimavanserin is administered at a dose of about 0.4 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.5 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 1 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 2 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 3 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 4 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 5 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 6 mg/kg.
  • pimavanserin is administered at a dose of about 7 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 8 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 9 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 10 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 25 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 30 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 50 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 60 mg/kg.
  • pimavanserin is administered at a dose of about 75 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 100 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 125 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 150 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 175 mg/kg. In some
  • pimavanserin is administered at a dose of about 200 mg/kg.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering doxepm, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
  • the present disclosure provides doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of doxepin, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of doxepin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is doxepin, or a pharmaceutically acceptable salt thereof.
  • doxepin is administered at a dose between about 0.010 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 0.05 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 0.1 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 0.5 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 45 mg/kg.
  • doxepin is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, doxepm is administered at a dose between about 1 mg/kg and about 35 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 30 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 25 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 20 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 15 mg/kg.
  • doxepin is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, doxepm is administered at a dose between about 5 mg/kg and about 25 mg/kg. In some embodiments, doxepm is
  • doxep is administered at a dose between about 5 mg/kg and about 20 mg/kg. In some embodiments, doxep is administered at a dose between about 5 mg/kg and about 15 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 30 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 25 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 20 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 15 mg/kg.
  • doxepin is administered at a dose of about 0.010 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.015 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.020 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.025 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.05 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.1 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.2 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.3 mg/kg.
  • doxepin is administered at a dose of about 0.4 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.5 mg/kg. In some embodiments, doxepin is administered at a dose of about 1 mg/kg. In some embodiments, doxepin is administered at a dose of about 2 mg/kg. In some embodiments, doxepin is administered at a dose of about 3 mg/kg. In some embodiments, doxepin is administered at a dose of about 4 mg/kg. In some embodiments, doxepin is administered at a dose of about 5 mg/kg. In some embodiments, doxepin is administered at a dose of about 10 mg/kg.
  • doxepin is administered at a dose of about 15 mg/kg. In some embodiments, doxepin is administered at a dose of about 20 mg/kg. In some embodiments, doxepin is administered at a dose of about 25 mg/kg. In some embodiments, doxepin is administered at a dose of about 30 mg/kg. In some embodiments, doxepin is administered at a dose of about 35 mg/kg. In some embodiments, doxepin is administered at a dose of about 40 mg/kg. In some embodiments, doxepin is administered at a dose of about 45 mg/kg. In some embodiments, doxepin is administered at a dose of about 50 mg/kg.
  • the compound is administered orally. In some embodiments, the compound is administered parenteralfy. In some embodiments, the compound is administered transderma!ly. In some embodiments, the compound is administered subcutaneously. In some embodiments, the compound is administered intravenously. [0278] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering diphenhydramine, or a
  • the present disclosure provides diphenhydramine, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of diphenhydramine, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of diphenhydramine, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is diphenhydramine, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the present disclosure provides gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
  • the present disclosure provides the use of gamma- hydroxybutyrate, for alleviating a symptom of, treating, or preventing a sleep disorder
  • the present disclosure provides the use of gamma- hydroxybutyrate, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
  • the compound is gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof
  • the compound of the present disclosure is not suvorexant, zolpidenr, S-zopiclone, or a pharmaceutically acceptable salt thereof.
  • the compound of the present disclosure is not suvorexant, or a pharmaceutically acceptable salt thereof.
  • the compound of the present disclosure is not zolpidem, or a pharmaceutically acceptable salt thereof.
  • the compound of the present disclosure is not S-zopiclone, or a pharmaceutically acceptable salt thereof.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respirator disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • RDI respirator disturbance index
  • PLMD periodic leg movement disorder
  • REM behavior disorder elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by sleep apnea, restless legs syndrome, a high respirator ⁇ ' disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • RDI respirator ⁇ ' disturbance index
  • PLMD periodic leg movement disorder
  • REM behavior disorder elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with sleep apnea, restless legs syndrome, high respiratory disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • RDI respiratory disturbance index
  • PLMD periodic leg movement disorder
  • REM behavior disorder elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by sleep apnea.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with sleep apnea.
  • sleep apnea is obstructive sleep apnea.
  • sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea.
  • RRI respiratory disturbance index
  • RERA elevated respiratory event related arousal
  • sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory' event related arousal (RERA) with a concomitant apnea.
  • RRI respiratory disturbance index
  • RERA elevated respiratory' event related arousal
  • sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant hypopnea.
  • RRI respiratory disturbance index
  • RERA elevated respiratory event related arousal
  • sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) wi th a concomitant hypopnea.
  • RRI respiratory disturbance index
  • RERA elevated respiratory event related arousal
  • sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without concomitant acute hemoglobin desaturation.
  • RRI respiratory disturbance index
  • RERA elevated respiratory event related arousal
  • sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with concomitant acute hemoglobin desaturation.
  • RRI respiratory disturbance index
  • RERA elevated respiratory event related arousal
  • sleep apnea is central sleep apnea.
  • sleep apnea is low-arousal threshold sleep apnea.
  • sleep apnea is hypopnea.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with restless legs syndrome.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by restless legs syndrome.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with restless legs syndrome.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a high respiratory disturbance index
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a high respiratory disturbance index (RDI).
  • RDI respiratory disturbance index
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a high respiratory disturbance index (RDI).
  • RDI respiratory disturbance index
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea.
  • RERA respiratory event related arousal
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with a concomitant apnea.
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with or without a concomitant hypopnea.
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with a concomitant hypopnea.
  • RERA respiratory event related arousal
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with or without concomitant acute hemoglobin desaturation.
  • RERA respiratory event related arousal
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with concomitant acute hemoglobin desaturation.
  • RERA respiratory event related arousal
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with or without concomitant hemoglobin desaturation.
  • RERA respiratory event related arousal
  • the RDI is associated with an elevated respiratory event related arousal (RERA) with concomitant hemoglobin desaturation.
  • RERA respiratory event related arousal
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a neurological disease.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a neurological disease.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a neurological disease.
  • the neurological disease is a neurodegenerative disease.
  • the neurodegenerative disease is Lewy body disease (i.e., Lewy body dementia). In some embodiments, the Lewy body disease is diffuse.
  • the neurodegenerative disease is amyotrophic lateral sclerosis (ALS). In some embodiments, the neurodegenerative disease is Huntington’s disease. In some embodiments, the neurodegenerative disease is Parkinson’s disease. In some embodiments, the neurodegenerative disease is Alzheimer’s disease. In some embodiments, the neurodegenerative disease is a synucleinopathy.
  • ALS amyotrophic lateral sclerosis
  • the neurodegenerative disease is Huntington’s disease.
  • the neurodegenerative disease is Parkinson’s disease.
  • the neurodegenerative disease is Alzheimer’s disease.
  • the neurodegenerative disease is a synucleinopathy.
  • a synucleinopathy is Alzheimer’s disease, Parkinson’s disease, or Lewy body dementia. In some embodiments, a synucleinopathy is Alzheimer’s disease. In some embodiments, a synucleinopathy is Parkinson’s disease. In some embodiments, a synucleinopathy is dementia with Lewy bodies. In some embodiments, a synucleinopathy is multiple system atrophy.
  • the neurological disease is a neurodevelopmental disease. In some embodiments, the neurodevelopmental disease is autism. In some embodiments, the neurological disease is a muscular dystonia. In some embodiments, the dystonia is neuromuscular dystonia. In some embodiments, the neuromuscular dystonia is spasmodic torticollis.
  • the neurological disease is multiple sclerosis (MS).
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a circadian rhythm disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a circadian rhythm disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a circadian rhythm disorder.
  • the circadian rhythm disorder is advanced sleep- wake phase disorder. In some embodiments, the circadian rhythm disorder is irregular sleep-wake rhythm disorder. In some embodiments, the circadian rhythm disorder is jet lag. In some embodiments, the circadian rhythm disorder is shift work sleep disorder. In some embodiments, the circadian rhythm disorder is delayed sleep phase syndrome. In some embodiments, the circadian rhythm disorder is non-24 hour rhythm disorder. [0335] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with pain.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by pain.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with pain.
  • the pain is selected from an inflammatory' pain, a nociceptive pain, a neuropathic pain, a mixed nociceptive and neuropathic pain, a post-operative pain, a post herpetic pain, a traumatic pain, a phantom-limb pain, a fibromyalgia, a back pain, a cancer pain, and an osteoart.hr itic pain.
  • the pain is an inflammatory pain.
  • the inflammatory pain is arthritis.
  • the arthritis is rheumatoid arthritis.
  • the arthritis is osteoarthritis.
  • the pain is a nociceptive pain.
  • the nociceptive pain is acute.
  • the nociceptive pain is chronic.
  • the nociceptive pain is caused by a cancer therapy.
  • the nociceptive pain is caused by a surgery.
  • the pain is a neuropathic pain.
  • the neuropathic pam is chronic.
  • the neuropathic pain is acute.
  • the neuropathic pam is back pam.
  • the neuropathic pam is caused by a spinal cord injury.
  • the neuropathic pain is caused by multiple sclerosis.
  • the neuropathic pam is caused by a stroke.
  • the neuropathic pain is caused by diabetes. In some embodiments, the neuropathic pain is caused by a metabolic condition.
  • the pain is a mixed nociceptive and neuropathic pain. [0343] In some embodiments, the pain is a post-operative pain.
  • the pain is a post-herpetic pain.
  • the pain is a traumatic pain.
  • traumatic pain is caused by causalgia.
  • the pam is a phantom-limb pain.
  • the pain is a fibromyalgia.
  • the pam is a back pam. In some embodiments, the pam is a low back pain.
  • the pam is a cancer pain.
  • the cancer pain is cancer.
  • the cancer pain is caused by a tumor.
  • the cancer pain is caused by a cancer treatment.
  • the cancer pain is caused by chemotherapy.
  • the cancer pain is radiation therapy.
  • the cancer pam is caused by surgery.
  • the pain is an osteoarthritic pain
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with periodic leg movement disorder
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by periodic leg movement disorder (PLMD).
  • PLMD periodic leg movement disorder
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with periodic leg movement disorder (PLMD).
  • PLMD periodic leg movement disorder
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with REM behavior disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by REM behavior disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with REM behavior disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with elderly fragmented sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by elderly fragmented sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with elderly fragmented sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with age-related sleep fragmentation.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by age-related sleep fragmentation.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with age-related sleep fragmentation.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with post-menopausal sleep disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by post-menopausal sleep disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with post-menopausal sleep disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with substance abuse.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by substance abuse.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with substance abuse.
  • the substance abuse is opioid abuse or alcoholism. In some embodiments, the substance abuse is opioid abuse. In some embodiments, the substance abuse is alcoholism.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with substance abuse withdrawal.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by substance abuse withdrawal.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with substance abuse withdrawal.
  • the substance abuse withdrawal is opioid withdrawal or alcohol withdrawal. In some embodiments, the substance abuse withdrawal is opioid withdrawal. In some embodiments, the substance abuse is alcohol withdrawal.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with narcolepsy.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject wherein the sleep disorder is caused by narcolepsy.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with narcolepsy.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a mental disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a mental disorder.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a mental disorder.
  • the mental disorder is depression, major depressive disorder, post- traumatic stress disorder, anxiety disorder, bipolar disorder, or schizophrenia.
  • the mental disorder is depression. In some embodiments, the mental disorder is major depressive disorder. In some embodiments, the mental disorder is post- traumatic stress disorder. In some embodiments, the mental disorder is anxiety disorder. In some embodiments, the mental disorder is bipolar disorder. In some embodiments, the mental disorder is schizophrenia.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with non-restorative sleep.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with snoring.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by snoring.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with snoring.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with idiopathic hypersomnia.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by idiopathic hypersomnia.
  • the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with idiopathic hypersomnia.
  • the number of arousals is decreased by up to about 99%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, about 50%, about 45%, about 40%, about 35%, about 30%, about 25%, about 20%, about 15%, about 10%, or about 5% reduction in the number of wake bouts per hour post-treatment relative to vehicle treatment.
  • gabalin alleviates a symptom of, treats, or prevents a sleep disorder.
  • gabapentin decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment. In some embodiments, gabapentin increases sleep consolidation as demonstrated by >2-foJd increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, gabapentin increases EEG slow' wave activity in nonREM sleep. [0394] In some embodiments, pregabalin alleviates a symptom of, treats, or prevents a sleep disorder.
  • pregabalin decreases the number of arousals (approximately 40% reduction in the number of wake bouts per hour post-treatment relative to vehicle treatment). In some embodiments, pregabalin increases sleep consolidation as evidenced by a nearly 3 -fold increase in average sleep bout duration per hour post-treatment relative to vehicle treatment.
  • mirogabaiin alleviates a symptom of, treats, or prevents a sleep disorder.
  • mirogabaiin decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment.
  • mirogabaiin increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, mirogabaiin increases EEG slow wave activity in nonREM sleep.
  • trazodone alleviates a symptom of, treats, or prevents a sleep disorder.
  • trazodone decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment.
  • trazodone increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment.
  • trazodone increases EEG slow wave activity in nonREM sleep.
  • nelotansenn alleviates a symptom of, treats, or prevents a sleep disorder.
  • nelotansenn decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment.
  • nelotansenn increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment.
  • neJotanserin increases EEG slow wave activity in nonREM sleep.
  • pimavanserin alleviates a symptom of, treats, or prevents a sleep disorder.
  • pimavanserin decreases arousals as demonstrated by a reduced number of w3 ⁇ 4ke bouts per hour post-treatment relative to vehicle treatment.
  • pimavanserin increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment.
  • pimavanserin increases EEG slow wave activity in nonREM sleep.
  • trazodone reduce sleep fragmentation.
  • nelotanserm reduce sleep fragmentation.
  • pimavanserin reduce sleep fragmentation.
  • trazodone decreases the number of arousals (measured by the number of wake bouts per hour post-treatment relative to vehicle treatment). In some embodiments, trazodone increases sleep consolidation.
  • nelotanserin decreases the number of arousals (measured by the number of w3 ⁇ 4ke bouts per hour post-treatment relative to vehicle treatment). In some embodiments, nelotanserin increases sleep consolidation (approximately 3 -fold increase m average sleep bout duration per hour post-treatment relative to vehicle treatment).
  • pimavanserin decreases the number of arousals (measured by the number of wake bouts per hour post-treatment relative to vehicle treatment).
  • pimavanserin increases sleep consolidation (as measured by average sleep bout duration per hour post-treatment relative to vehicle treatment).
  • the efficacy achieved with pimavanserin may be less than that observed for trazodone and nelotanserin.
  • the comparatively smaller efficacy of pimavanserin on sleep consolidation may be due to an efficacy ceiling (maximum achievable magnitude of effect) as stepwise dose escalation up to 60 mg/kg produced no further increase in average sleep bout duration per hour.
  • doxepin alleviates a symptom of, treats, or prevents a sleep disorder.
  • doxepin reduces sleep fragmentation.
  • doxepin decreases the number of arousals (measured by the number of w3 ⁇ 4ke bouts per hour post-treatment relative to vehicle treatment). In some embodiments, doxepin increases sleep consolidation (approximately 2-fold increase in average sleep bout duration per hour post-treatment relative to vehicle treatment).
  • diphenydramine alleviates a symptom of, treats, or prevents a sleep disorder.
  • diphenydramine decreases arousals as demonstrated by a reduced number of w3 ⁇ 4ke bouts per hour post-treatment relative to vehicle treatment.
  • diphenydramine increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment.
  • diphenydramine increases EEG slow wave activity in nonREM sleep.
  • gamma-hydroxybutyrate alleviates a symptom of, treats, or prevents a sleep disorder.
  • gamma-hydroxybutyrate decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment.
  • gamma-hydroxybutyrate increases sleep consolidation as demonstrated by an increase m duration of average sleep bouts per hour post-treatment relative to vehicle treatment.
  • gamma-hydroxybutyrate increases EEG slow 7 wave activity 7 in nonREM sleep.
  • one or more compounds of the present disclosure produces improvements in sleep fragmentation in comparison to compounds prescribed comprising the pharmacological standard of care for insomnia.
  • the standard of care compounds for insomnia are, for example, but not limited to, the orexin antagonists (e.g., suvorexant (BELSOMRA®)) and non-benzodiazepine, benzodiazepine receptor dependent GABAA allosteric modulators (e.g., zolpidem (AMB1EN®) and eszopiclone (LUNESTA®)), which do not adequately improve the requisite precimical measures of sleep fragmentation.
  • the orexin antagonists e.g., suvorexant (BELSOMRA®)
  • non-benzodiazepine benzodiazepine receptor dependent GABAA allosteric modulators
  • benzodiazepine receptor dependent GABAA allosteric modulators e.g., zolpidem (AMB1EN®) and eszopiclone (LUNESTA
  • orexin OC1 / ⁇ C2 antagonist suvorexant fails to improve sleep fragmentation at 3 mg/kg, 10 mg/kg, and 30 mg/kg.
  • suvorexant does not reduce the number of arousals as measured by the number of wake bouts per hour, but instead, increases the number of arousals.
  • suvorexant produces small
  • zolpidem at doses sufficient to produce increases m soporific efficacy as measured by EEG slow wave activity (EEG delta power in nonREM sleep), fails to reduce the number of arousals as measured by the number of wake bouts per hour and fails to produce improvement in sleep consolidation as measured by average sleep bout duration per hour.
  • Zolpidem can produce sedative effects (up to and including non-responsiveness of the subject); however, increasing the dose of zolpidem may be complicated by a short kinetic half- life and can be contraindicated due to significant unwanted side effects including, but not limited to, ITEM sleep inhibition, severe motor coordination impairment, memory impairment, rebound insomnia, and other unwanted adverse effects, or a combination thereof.
  • S-zopiclone (marketed as LUNESTA®) fails to reduce the number of arousals as measured by the number of wake bouts per hour and produces only small improvements in sleep consolidation as measured by average sleep bout duration per hour.
  • the dose of S-zopiclone tested i.e., from about 5 mg/kg to about 30 mg/kg
  • S- zopiclone decreases Locomotor Activity Intensity (LMAi).
  • S-zopiclone dose-dependently inhibits REM sleep, which may be important for memory and learning.
  • standard of care insomnia drugs e.g., zolpidem and S-zopiclone
  • relaxation of upper airway muscles is undesirable as it could worsen obstructive sleep apnea and its comorbid health risks.
  • Direct electromyographic (EMG) assessment of muscle tone may reveal unwanted myorelaxation (decrease in skeletal muscle electrical activity) after treatment with S-zopiclone.
  • EMG Direct electromyographic
  • Example 1 Experimental details and methods for determining sleep continuity
  • cyanoacrylate applied between the hermetically sealed implant connector and skull, and dental acrylic.
  • An analgesic (buprinorphine 0.3 mg/kg IP) was administered pre-operatively and daily SC for 2 days post-surgery.
  • An antibiotic was administered before surgery (chloramphenicol 40 mg/kg IM) and for 7-10 days after surgery (Clavamox b.i.d). At least three weeks were allowed for surgical recovery prior to any data collection.
  • Rats were housed individually within specially modified Nalgene ® microisolator cages equipped with an ultra-low-torque slip-ring commutator and a custom polycarbonate filter-top riser. These cages were located within separate, ventilated compartments of a stainless steel sleep-wake recording chamber. Food and water were available ad libitum and the ambient temperature was 23 ⁇ 1° C. A 24-hr light-dark cycle (LD 12: 12) was maintained throughout the study using fluorescent light. Light intensity averaged 35-40 lux at mid-level inside the cage. Relative humidity averaged 50% approximately. Animals were undisturbed for two days before and after each treatment.
  • Arousal states were classified on-line as NREM sleep, REM sleep, wake, or theta-dominated wake every 10 seconds using EEG period and amplitude feature extraction and ranked membership algorithms.
  • Individually taught EEG-arousal-state templates and EMG criteria differentiated states of arousal.
  • LMA and drink-related activity' were automatically recorded as counts per minute, and body temperature was recorded each minute.
  • LMA was detected in both horizontal and vertical planes by a customized telemetry' receiver (ER4000, Minimitter, Bend, OR) beneath the cage.
  • Drink-related activity' and food-related activity' were detected by beam break sensors closely situated around recessed access portals to the lixit and the food bin, respectively.
  • the beam break area for the food bin was, however, relatively large, and these data have not been validated as an endpoint for food consumpti on per se.
  • Telemetry measures (LMA and body temperature) were not part of the SCORE arousal -state determination algorithm; thus, sleep-scoring and telemetry data were concurrent but independent measures.
  • methy!ee!lulose vehicle was prepared as a sterile 0.25% solution of methylcellulose (15 centipoise, Sigma, St. Louis, MO., USA).
  • HPitCD (American Maise Products, Indianapolis, IN) vehicle was initially prepared as a 50% stock solution (w/v in 0.9% NaCl) and then further diluted with 0.9% NaCl to achieve percent solutions as reported for respective studies.
  • Intraperitoneal administration was delivered at a volume of 1 mL/kg
  • Oral dosing was at a volume of 1 -2 mL/kg.
  • the standard recording duration for SCORE data was not less than 30 hours before and after treatment.
  • the 30 hours pre-treatment baseline recording was itself preceded by at least 24 hours in which the animal was undisturbed in the home/recording cage.
  • Rats were randomly assigned to treatments in parallel groups. Some rats received more than one active treatment, in which cases at least 7 days“washout” elapsed between each treatment.
  • Post-treatment detail plots the first five hours post- treatment. Variables were computed in 5-minute bins, aligned to the minute of treatment. The first time bin, labeled 0, represents the first 5 minutes post treatment.
  • Pre- and post-treatment time series plot ⁇ 30 hours before and after CT-18 treatments (or 29 hr before and 31 hr after CT-5 treatments). Treatment occurred at the beginning of the hour marked by an arrow. Variables were computed in hourly bins.
  • Gabapentin reduces number of arousals
  • Gabapentin produces robust increases in sleep continuity/consolidation
  • EEG slow wave activity is indicative of increased arousal threshold and depth of sleep due to the soporific effects of gabapentin (FIG. 3).
  • Example 3 Effects of pregabalin on number of arousals and sleep continuity
  • Pregahalin reduces number of arousals
  • a2d-1 / a2d ⁇ 2 ligand pregabalin administered to Wistar rats at CT ⁇ 1 8 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), significantly decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts per hour, as compared to vehicle control treatment. Number of wake bouts are plotted as hourly mean ⁇ SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 4).
  • Pregabalin increases sleep continuity/consolidation
  • a2d-!/ a2d-2 ligand pregabalin administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation as demonstrated by an >2-fold increase in average sleep bout duration per hour post-treatment (as indicated by the arrow) relative to vehicle control treatment.
  • the robust sleep consolidating effect of pregabalin was approximately 2 times greater than the circadian peak for this measure during the undisturbed baseline circadian rest phase (circadian time 0:00-12:00).
  • Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated m each hour for an individual animal, plotted as the population hourly mean ⁇ SEM 30 hours before (baseline) and after treatment. Group sizes (N) are shown in the graph legend.
  • Statistically significant differences from methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 5).
  • Trazodone reduces number of arousals
  • the SHTIA antagonist trazodone administered to Wistar rats at CT-18 (6 hours after light- off; time of treatment indicated by the triangle on the abscissa), decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts, as compared to vehicle control treatment. Number of wake bouts are plotted as hourly mean ⁇ SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 6).
  • Trazodone increases sleep continuity/ consolidation
  • Example 5 Effects of nelotanseriii on number of arousals and sleep continuity
  • Nelotanserin reduces number of arousals
  • the 5HT2A antagonist/inverse agonist nelotansenn administered to Wistar rats at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts, as compared to vehicle control treatment.
  • Number of wake bouts are plotted as hourly mean ⁇ SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylceilulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 8).
  • Nelotanserin increases sleep continuity/consolidation
  • Example 6 Effects of pimavanserin on number of arousals and sleep continuity
  • Pimavanserin reduces number of arousals
  • methylceilulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 10).
  • Pimavanserin increases sleep continuity/consolidation
  • Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in a given hour for an individual animal, plotted as the group hourly mean ⁇ SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 11).
  • Example 7 Effects of doxepm on number of aronsals and sleep continuity
  • Doxepin reduces number oj iron sals
  • Doxepin increases sleep continuity /consolidation
  • Example 8 Effects of suvorexant on number of arousals and sleep continuity
  • Example 9 Effects of zolpidem on soporific efficacy, number of arousals, sleep continuity, REM sleep, and locomotor activity
  • methylcellulose vehicle control are indicated by asterisks.
  • 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 16).
  • methylcellulose vehicle control are indicated by asterisks.
  • 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 18).
  • methylcellulose vehicle control are indicated by asterisks.
  • 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 19).
  • Example 10 Effects of S-zopiclone on number of arousals, sleep continuity, locomotor activity, muscle tone, and rebound sleep disturbance
  • Locomotor Activity Intensity (LMAi) as measured by the counts of locomotor activity per minute of wakefulness, averaged hourly for each animal.
  • LMAi reveals whether changes in motor activity (movements) are disproportionate from that normally observed during wakefulness. A disproportionate reduction in locomotor activity during wakefulness is shown. The reduction in LMAi increases with dose and is evident 1-6 hours post-treatment in the S- zopiclone 10 mg/kg and 30 mg/kg example (as indicated by the arrow).
  • EMG electromyogram

Abstract

The present disclosure relates to methods of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of α2δ-1 auxiliary subunit of voltage-gated calcium channels, a ligand of α2δ-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist, a H1 receptor antagonist, a H1 receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or pharmaceutically acceptable salts thereof, to a subject in need thereof.

Description

METHODS FOR TREATING SLEEP FRAGMENTATION DISORDERS
RELATED APPLICATION
[0001] This application claims priority to, and the benefit of U.S. Provisional Application No. 62/815,610, filed March 8, 2019, the entire contents of which are incorporated by reference.
FIELD OF THE APPLICATION
[0002] The present disclosure relates to methods of alleviating a symptom of, treating, or preventing a sleep disorder (e.g., increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold) by administering a compound of the present disclosure, or pharmaceutically acceptable salt thereof.
BACKGROUND
[0003] Breakthroughs m the field of sleep disorders research has brought about widespread scientific and popular appreciation for the health benefits of restful and restorative sleep. Sleep is now' recognized along with diet and exercise as one of the three pillars of good health. The overall prevalence of current or previous sleep disorders in adults is estimated at 52.4% of the population. Almost two-thirds (64%) of the population report sleep difficulties at least a few times a week (National Sleep Foundation,“2012 Sleep m America” Poll). The International Classification of Sleep Disorders distinguishes over 80 different disorders and each can have profound health and economic implications. Whilst the daytime impairment caused by poor sleep has long been appreciated, poor sleep also has cascading negative impact upon alertness, cognition, learning and memory, vigilance, performance, and a broad range of co-morbid health conditions including acute and chronic pam and pain disorders, psychiatric conditions, neurodegenerative disease, developmental disorders, metabolic disease and diabetes, obesity , cardiovascular disease, immunological disorders, and many other medical conditions.
[0004] Sleep disorder patients are now readily segmented into a broader range of sleep disorder categories and conditions that are more amenable to new and better tailored therapies that hold promise for delivering better patient outcomes. Objective measures of sleep can play a vital role toward understanding poor sleep and its amelioration. Despite sleeping 7-8 hours or more, patients whose sleep is frequently interrupted or“fragmented”, suffer all the consequences of sleep deprivation. Sleep consolidation is necessary for the restorative physiological benefits of sleep to be realized and comorbid conditions to be managed.
[0005] Pharmacological options are limited for the treatment of sleep fragmentation. There exists an urgent unmet clinical need to develop new methods of treating sleep fragmentation, including pharmacological methods of treatment.
SUMMARY
[0006] In one aspect, the present disclosure provides compounds for alleviating a symptom of, treating, or preventing a sleep disorder.
[0007] In some embodiments, the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold. In some embodiments, the sleep disorder is increased disturbed sleep. In some embodiments, the sleep disorder is increased sleep fragmentation. In some embodiments, the sleep disorder is increased arousals. In some embodiments, the sleep disorder is decreased arousal threshold.
[0008] Compounds of the present di sclosure were studied for their effects on EEG sleep- wakefulness, locomotor activity, drink- and food-related activity and body temperature in laboratory rats using an enhanced and expanded version of SCORE-2000®, a sleep-wake bioassay and analysis system known as“SCORE™”.
[0009] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAs receptor agonist.
[0010] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a2d-1 and a2d-2 auxiliary subunits of voltage-gated calcium channels.
[001 1] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels. [0012] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a ligand of a a2d-2 auxiliar subunit of voltage-gated calcium channels.
[0013] in one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a 5HT:?A receptor antagonist and inverse agonist.
[0014] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wlierein the compound is a 5HT2A receptor antagonist or a 5HT2A receptor inverse agonist.
[0015] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a 5HT2A receptor antagonist.
[0016] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a 5HT2A receptor inverse agonist.
[0017] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor antagonist and inverse agonist.
[0018] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor antagonist or a Hi receptor inverse agonist.
[0019] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor antagonist.
[0020] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a Hi receptor inverse agonist.
[0021 ] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a gamma-hydroxy butyrate (GHB) receptor ligand.
[0022] In one aspect, the present disclosure provides the identification of a pharmacological compound class, wherein the compound is a GABAB receptor agonist.
[0023] In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage gated calcium channels is selected from gabapentm, pregabalm, and mirogabalin.
[0024] In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage gated calcium channels is selected from gabapentm and pregabalin.
[0025] In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage gated calcium channels is gabapentin. In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage-gated calcium channels is pregabalin. In some embodiments, the ligand of an a2d-1 and a2d-2 auxiliary subunit of voltage-gated calcium channels is mirogabalin.
[0026] In some embodiments, the 5HT?.A receptor antagonists and inverse agonists is selected from trazodone, ne!otanserin, and pimavanserin.
[0027] In some embodiments, the 5HT?.A receptor antagonist or 5HT:?A receptor inverse agonist is selected from trazodone, nelotanserin, and pimavanserin.
[0028] In some embodiments, the Hi receptor antagonist and inverse agonist is selected from doxepm and diphenhydramine.
[0029] In some embodiments, the Hi receptor antagonist or Hi receptor inverse agonist is selected from doxepin and diphenhydramine.
[0030] In some embodiments, the Hi receptor antagonist and inverse agonist is doxepin.
[0031 ] In some embodiments, the Hi receptor antagonist or Hi receptor inverse agonist is doxepin.
[0032] In some embodiments, the gamma-hydroxy butyrate (GHB) receptor ligand is gamma- hydroxybutyrate.
[0033] In some embodiments, the GABAB receptor agonist is gamma-hydroxybutyrate.
[0034] In some embodiments, the gamma-hydroxybutyrate is the sodium or potassium salt.
[0035] In some embodiments, a compound of the present disclosure is selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
[0036] In some embodiments, a compound of the present disclosure is selected from gabapentin, pregabalin, trazodone, nelotanserin, pimavanserin, doxepin, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
[0037] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds of the present disclosure selected from gabapentin, pregabalin, mirogabalin, trazodone,
nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof to a subject in need thereof.
[0038] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering gabapentin, or a pharmaceutically acceptable salt thereof. [0039] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering pregabalin, or a pharmaceutically acceptable salt thereof.
[0040 ] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering mirogabalin, or a pharmaceutically acceptable salt thereof.
[0041] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering trazodone, or a pharmaceutically acceptable salt thereof.
[0042] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering nelotanserin, or a pharmaceutically acceptable salt thereof
[0043] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering pimavanserin, or a pharmaceutically acceptable salt thereof.
[0044] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering doxepin, or a
pharmaceutically acceptable salt thereof
[0045] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering diphenhydramine, or a pharmaceutically acceptable salt thereof.
[0046] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
[0047] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respiratory disturbance index (RD1), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
[0048] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea.
[0049] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with restless legs syndrome.
[0050] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a high respiratory disturbance index (RDI).
[0051] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a neurological disease.
[0052] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a circadian rhythm disorder.
[0053] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with pain.
[0054] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with periodic leg movement disorder
(PLMD).
[0055] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with REM behavior disorder.
[0056] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with elderly fragmented sleep.
[0057] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with age-related sleep fragmentation.
[0058] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with post-menopausal sleep disorder.
[0059] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-rnorbid with substance abuse.
[0060] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with substance abuse withdrawal.
[0061] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with narcolepsy. [0062] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a mental disorder.
[0063] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with non-restorative sleep.
[0064] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with snoring.
[0065] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with idiopathic hypersomnia.
[0066] In some embodiments, the methods of the present disclosure the subject is administered a pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises a compound of the present disclosure and an additional active agent. In some embodiments, the additional active agent is a sedative-hypnotic.
[0067] In some embodiments, the sedative-hypnotic is selected from zolpidem, suvorexant, butabarbitai, quazepam, triazolam, tasimelteon, eszopiclone, temazepam, ramelteon,
secobarbital, doxepin, estazolam, and flurazepam.
[0068] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder by administering a pharmaceutical composition comprising a ligand of a2d-1 and a2d-2 auxiliary subunits of voltage-gated calcium channels, a 5HT2A receptor antagonists, a 5HT2A receptor inverse agonists, a Hi receptor antagonists, a Hi receptor inverse agonists, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or a pharmaceutically acceptable derivative thereof, in combination with an additional active agent. [0069] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder by administering a pharmaceutical composition comprising a compound of the present disclosure, or a pharmaceutically acceptable derivative thereof, in combination with one or more additional active agent.
[0070] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder by administering the compounds of the present disclosure and any additional active agents, if present, either horn somni, h.s. (at bedtime) or between about 0-4 hours before bedtime to a subject m need thereof.
[0071] Pharmaceutical therapies and methods of using such pharmaceutical therapies are provided herein for treating a sleep disorder. Pharmaceutical therapies and methods of using such pharmaceutical therapies are provided for a sleep disorder in a subject with one or more co- morbid medical conditions. In some embodiments, these therapies and methods of using such therapies include pharmaceutical therapies to decrease disturbed sleep, decrease sleep fragmentation, decrease arousals, or increase arousal threshold. In some embodiments, a subject may have one or more of the following medical conditions: sleep apnea, restless legs syndrome, a high respiratory disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
[0072] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting.
[0073] Other features and advantages of the disclosure will be apparent from the following detailed description and claims. BRIEF DESCRIPTION OF THE FIGURES
[0074] FIG. 1 depicts the number of wake bouts each hour after treatment with gabapentin, wherein the thin line depicts gabapentin administration at 100 mg/kg PO (CT-5, n=9), and the thick line depicts methylcellulose administration at 1 niL/kg PO (CT-5, n=21).
[0075] FIG. 2 depicts the average aligned sleep bout each hour after treatment with gabapentin, wherein the thin line depicts gabapentin administration at 100 mg/kg PO (CT-5, n=9), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-5, n=21).
[0076] FIG. 3 depicts normalized EEG delta power after treatment with gabapentin, wherein the thin line depicts gabapentin administration at 100 mg/kg PO (CT-5, n=9), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-5, n=21).
[0077] FIG. 4 depicts the number of wake bouts each hour after treatment with pregabaiin, wherein the thin line depicts pregabaiin administration at 10 mg/kg PO (n=8), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=I0)
[0078] FIG. 5 depicts the average sleep bout each hour after treatment with pregabaiin, wherein the thin line depicts pregabaiin administration at 10 mg/kg PO (n=8), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=10).
[0079] FIG. 6 depicts the number of wake bouts each hour after treatment with trazodone, wherein the thin line depicts trazodone administration at 30 mg/kg PO (CT-18, n=9), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-1 8, n=T0).
[0080] FIG. 7 depicts the average sleep bout each hour after treatment with trazodone, wherein the thin line depicts trazodone administration at 20 mg/kg PO (CT-18, n=T 2), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=::12).
[0081] FIG. 8 depicts the number of wake bouts per hour after treatment with nelotanserin, wherein the thin line depicts nelotanserin administration at 10 mg/kg PO (CT-5, n:=:12), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-5, n=T 5).
[0082] FIG. 9 depicts the average aligned sleep bout each hour after treatment with nelotanserin, wherein the thin line depicts nelotanserin administration at 10 mg/kg PO and methylcellulose PO (CT-18, n==:8), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18 twice, n=8). [ 0083] FIG. 10 depicts the number of wake bouts per hour after treatment with pimavanserin, wherein the thin line depicts pimavanserin administration at 10 mg/kg PO (CT-5, n=l 1 ), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-5, n=12).
[0084] FIG. 11 depicts the averaged aligned sleep bout each hour after treatment with pimavanserin, wherein (A) depicts pimavanserin administration at 60 mg/kg PO (CT-18, n=12); (B) depicts pimavanserin administration at 30 mg/kg PO (CT-18, n=T0); (C) depicts
pimavanserin administration at 10 mg/kg PO (CT-18, n=10): and (H) depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=30).
[0085] FIG. 12 depicts the number of wake bouts each hour after treatment with doxepin, wherein the thin line depicts doxepin administration at 25 mg/kg PO (n=T7), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=T7).
[0086] FIG. 13 depicts the average sleep bout each hour after treatment with doxepin, wherein the thin line depicts doxepin administration at 25 mg/kg PO (n=I7), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=17).
[0087] FIG. 14 depicts the number of wake bouts each hour after treatment with suvorexant, wherein the thin line depicts suvorexant administration at 30 mg/kg PO (CT-18, n=7), and the thick line depicts TPGS (HY-16388) administration at 1 mL/kg PO (CT-18, n=9).
[0088] FIG. 15 depicts the average aligned sleep bout each hour after treatment with suvorexant, wherein the thin line depicts suvorexant administration at 30 mg/kg PO (CT-18, n=7), and the thick line depicts TPGS (HY-16388) administration at 1 mL/kg PO (CT-18, n=9).
[0089] FIG. 16 depicts the normalized EEC delta power after treatment with zolpidem tartrate, wherein the thin line depicts zolpidem tartrate administration at 10 mg/kg PO (CT-1 8, EEG telemetry, n=8), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, EEG telemetry , n=9).
[0090] FIG. 17 depicts the number of wake bouts each hour after treatment with zolpidem tartrate, wherein the thin line depicts zolpidem tartrate administration at 10 mg/kg PO (CT-18, EEG telemetry, n:=:8), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, EEG telemetry, n=9).
[0091] FIG. 18 depicts the average aligned sleep bout each hour after treatment with zolpidem tartrate, wherein the thin line depicts zolpidem tartrate administration at 10 mg/kg PO (CT-18, EEG telemetry, n=8), and the thick line depicts methylcellulose administration at 1 ml. kg PO (CT-18, EEG telemetry, n=9).
[0092] FIG. 19 depicts REM sleep after treatment with zolpidem, wherein the thin line depicts zolpidem administration at 30 mg/kg PO (n=T6), and the thick line depicts methylcellulose administration at 1 niL/kg PO (CT-5, n=16).
[0093] FIG. 20 depicts Locomotor Activity Intensity (LMAi) after treatment with zolpidem, wherein the thin line depicts zolpidem administration at 30 mg/kg PO (CT-18, n=10), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=10).
[0094] FIG. 21 depicts the number of wake bouts each hour after treatment with S-zopiclone, wherein the thin line depicts S-zopiclone administration at 10 mg/kg PO (CT-18, n=13), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=T3).
[0095] FIG. 22 depicts the average sleep bout each hour after treatment with S-zopiclone, wherein (A) depicts S-zopiclone administration at 30 mg/kg PO (CT-18, n=I2); (B) depicts S~ zopiclone administration at 10 mg/kg PO (CT-18, n=10); (C) depicts S-zopiclone administration at 5 mg/kg PO (CT-18, n=I2); and (D) depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=12).
[0096] FIG. 23 depicts the Locomotor Activity Intensity (LMAi) after treatment with S- zopiclone, wherein (A) depicts S-zopiclone administration at 30 mg/kg PO (CT-18, n=12); (B) depicts S-zopiclone administration at 10 mg/kg PO (CT-18, n=T0); (C) depicts S-zopiclone administration at 5 mg/kg PO (CT-18, n=T2); and (D) depicts methylcellulose administration at 1 mL/kg PO (CT-1 8, n 12).
[0097] FIG. 24 depicts the integrated EMG activity after treatment with S-zopiclone, wherein the thin line depicts S-zopiclone administration at 10 mg/kg PO (CT-18, n=T 3), and the thick line depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=T2).
[0098] FIG. 25 depicts the nonREM (NREM) sleep with S-zopiclone, wherein (A) depicts S- zopiclone administration at 30 mg/kg PO (CT-18, n=T2); (B) depicts S-zopiclone administration at 10 mg/kg PO (CT-18, n=T0); (C) depicts S-zopiclone administration at 5 mg/kg PO (CT-18, n=T2); and (D) depicts methylcellulose administration at 1 mL/kg PO (CT-18, n=T2). DETAILED DESCRIPTION
Definitions
[0099] As used herein,“uninterrupted bouts of sleep” (average sleep bout duration) refer to the average duration of all bouts of uninterrupted sleep that occurred each hour, measured in minutes.“Interruption” of sleep refers to 2 or more consecutive 10 second epochs of
wakefulness.“Interruption” of wake refers to 2 or more consecutive 10 second epochs of sleep. The value for the length of a bout that extends into the subsequent hour is assigned to the hour in which it begins. Analogous quantification is carried out for bouts of wakefulness. Sleep bout length may reflect the human tendency to awaken periodically through the night (such awakenings are normally not recalled), which in turn may be an important factor determining the restorative value of sleep in humans. Pre-eiinicai measures of sleep bout duration are also predictors of soporific efficacy in a subject (e.g., human).
[0100] As used herein,“NREM” and“nonREM” refer to non-rapid eye movement sleep stages.
[0101] As used herein,“reduced number of arousals” refers to the reduced number of wake bouts per hour.
[0102] As used herein,“REM” refers to the rapid eye movement sleep stage.
[0103] As used herein,“SEAT’ refers to standard error of the mean.
[0104] As used herein,“CT” refers to circadian time.
[0105] As used herein“sleep consolidation” refers to the measurement of the average sleep bout duration per hour.
[0106] As used herein,“sleep continuity" refers to the measurement of sleep-bout length.
[0107] As used herein, the“depth” of sleep is characterized by EEG slow wave activity, which may subserve sleep continuity or sleep consolidation, which is one of several determinants of sleep quality.
[0108] As used herein,“SWA” refers to slow wave activity, which may be exemplified as EEG delta po wer by use of Fourier analysis.
[0109] As used herein,“LMA intensity” (LMAi) refers to locomotor activity (LMA) counts per minute of EEG-defmed wakefulness. This variate allows an assessment of LMA that is independent of the amount of time awake, which may be used to quantify the specificity of a wake- or sleep-promoting effect. [0110] As used herein,“number of wake bouts” refers to the number of uninterrupted bouts of wakefulness that occurred each hour, measured in minutes. Number of wake bouts are of interest because it may closely reflects the number of arousals from sleep that occur at the time of measurement. Interpreted together, average sleep bout duration and number of wake bouts provide a highly reliable assessment of a drug or novel molecular entity (NME) effect on sleep fragmentation. Drugs that improve sleep fragmentation have been shown to improve the restorative benefits of sleep.
[0111] As used herein, the term“co-morbid” refers to a disease or disorder which is concurrent with a sleep disorder, but may not be the cause, in whole or in part, of the sleep disorder.
[0112] As used herein, the term“pharmaceutically acceptable” refers to compounds, anions, cations, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0113] It is to be understood that the present disclosure also provides pharmaceutical compositions comprising any compound described herein in combination with at least one pharmaceutically acceptable excipient or carrier.
[0114] As used herein, the term“pharmaceutical composition” is a formulation comprising a compound of the present disclosure in a form suitable for administration to a subject. In some embodiments, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial. The quantity of active ingredient (e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the subject. The dosage will also depend on the route of administration. A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, mtrapentoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for the topical or transdermal administration of a compound of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In some embodiments, the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
[0115] As used herein, the term“preventing,”“prevent,” or“protecting against” describes reducing or eliminating the onset of the symptoms or complications of such disease, condition or disorder.
[0116] It is to be appreciated that references to“treating” or“treatment” include the alleviation of established symptoms of a condition.“Treating” or“treatment” of a state, disorder or condition therefore includes: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclimcal symptoms of the state, disorder or condition, (2) inhibiting the state, disorder or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof (in case of maintenance treatment) or at least one clinical or subciinical symptom thereof! or (3) relieving or attenuating the disease, i.e., causing regression of the state, disorder or condition or at least one of its clinical or subciinical symptoms.
[0117] As used herein, the term“rat” is used interchangeably with“laboratory rat”.
[0118] As used herein, the term“subject” is interchangeable with the term“subject m need thereof’, both of which refer to a subject having a disease or having an increased risk of developing the disease. A“subject” includes a mammal. The mammal can be , for example, a human or appropriate non-human mammal, such as primate, mouse, rat, dog, cat, cow, horse, goat, camel, sheep or a pig. The subject can also be a bird or fowl. In some embodiments, the mammal is a rat. In some embodiments, the mammal is a human. In some embodiments, a subject in need has been previously diagnosed or identified as having a disease or disorder disclosed herein. A subject in need thereof can also be one who is suffering from a disease or disorder disclosed herein. Alternatively, a subject in need thereof can be one who has an increased risk of developing such disease or disorder relative to the population at large (i.e., a subject who is predisposed to developing such disorder relative to the population at large). A subject in need thereof can have a refractory or resistant a disease or disorder disclosed herein (i.e., a disease or disorder disclosed herein that does not respond or has not yet responded to treatment). The subject may he resistant at start of treatment or may become resistant during treatment. In some embodiments, the subject in need thereof received and failed all known effective therapies for a disease or disorder disclosed herein. In some embodiments, the subject in need thereof received at least one prior therapy.
[0119] Unless explicitly indicated otherwise, the terms“approximately” and“about” are synonymous. In some embodiments,“approximately” and“about” refer to the recited value, amount, dose, or duration ± 20%, ± 15%, ± 10%, ± 8%, ± 6%, ± 5%, ± 4%, ± 2%, ± 1%, or ± 0.5%. In another embodiment,“approximately” and“about” refer to the listed value, amount, dose, or duration ± 10%, ± 8%, ± 6%, ± 5%, ± 4%, or ± 2%. In some embodiments,
“approximately” and“about” refer to the listed value, amount, dose, or duration ± 5%. In some embodiments,“approximately” and“about” refer to the listed value, amount, dose, or duration ± 2%. In some embodiments,“approximately” and“about” refer to the listed amount, value, amount, dose, or duration ± 1%.
[0120] In some embodiments, a“compound” is the same as an“active ingredient”,“a ligand of «25-1 and «25-2 auxiliary' subunits of voltage-gated calcium channels, a 5HT2A receptor antagonists, a 5HT2A receptor inverse agonists, a Hi receptor antagonists, a Hi receptor inverse agonists, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAs receptor agonist”,“a ligand of «25-1 and «25-2 auxiliary' subunits of voltage-gated calcium channels”,“a 5HT2A receptor antagonists”,“a 5HT2A receptor inverse agonists”,“a Hi receptor antagonists”,“a Hi receptor inverse agonists”,“gamma-hydroxy butyrate (GHB) receptor ligand”,“a GABAs receptor agonist”, and a“compound of the present disclosure”.
[0121 ] In some embodiments,“gabapentin” may also be referred to as HY-10310 or 2-[l-
(aminomethyl)cyclohexyl]acetic acid
Figure imgf000017_0001
pharmaceutically acceptable salt thereof.
[0122] In some embodiments,“pregabaiin” may also be referred to as HY- 10204 or (3S)-3-
(aminomethyl)-5-methylhexanoic acid (i.e.,
Figure imgf000017_0002
or a pharmaceutically acceptable salt thereof. [0123] In some embodiments,“mirogabalin” may also be referred to as 2-[(lR,5S,6S)-6-
(aminomethyl)-3-ethyl-6-bicyclo[3.2.0]hept-3-enyl]acetic acid
Figure imgf000018_0001
pharmaceutically acceptable salt thereof.
[0124] In some embodiments,“trazodone” may also be referred to as 2- {3-[4-(3- chlorophenyl)piperazm- 1 -yl]propyl} -2H,3H- [ 1 ,2,4]triazolo[4,3 -a]pyndin-3 -one (i. e. ,
Figure imgf000018_0002
pharmaceutically acceptable salt thereof.
[0125] In some embodiments,“nelotansenn” may also be referred to as 3-[3-(4-bromo-l-methyl- lH-pyrazol-5-yl)-4-methoxyphenyl]-l-(2,4-difluorophenyl)urea (i.e.,
Figure imgf000018_0003
pharmaceutically acceptable salt thereof.
[0126] In some embodiments,“pimavanserm” may also be referred to as l-[(4- fluorophenyl)methyl]-l-(l -methylpipendin-4-yl)-3- {[4-(2-methylpropoxy)phenyl]methyl}urea
Figure imgf000018_0004
pharmaceutically acceptable salt thereof [0127] In some embodiments,“doxepm” may also he referred to as 3-(6H-
benzo[c] [ 1 ]benzoxepin- 11 -ylidene)-N,N-dimethylpropan- 1 -amine
Figure imgf000019_0001
pharmaceutically acceptable salt thereof
[0128] In some embodiments,“diphenhydramine” may also be referred to as 2-(benzhydryloxyj-
N,N-dimethylethan- 1 -amine
Figure imgf000019_0002
pharmaceutically acceptable salt thereof.
[0129] In some embodiments,“gamma-hydroxybutyrate” may also he referred to as 4-
0
hydroxybutanoic acid (i.e.,
Figure imgf000019_0003
or a pharmaceutically acceptable salt thereof.
[0130] In some embodiments, gamma- hydroxy butyrate is a pharmaceutically acceptable salt.
[0131] In some embodiments, gamma-hydroxybutyrate is the sodium salt.
[0132] In some embodiments, gamma-hydroxybutyrate is the potassium salt.
[0133] In some embodiments,“zolpidem” may also be referred to as Ambien®, HY- 10131, N,N-dimethyl-2-[6-methyl-2-(4-methylphenyl)imidazo[l,2-a]pyridin-3-yl]acetamide (i.e.,
Figure imgf000019_0004
pharmaceutically acceptable salt thereof.
[0134] In some embodiments, zolpidem is a pharmaceutically acceptable salt.
[0135] In some embodiments, zolpidem is zolpidem tartrate. [0136] In some embodiments,“S-zopiclone” may also be referred to as Lunesta®, eszopiclone, [(7S)-6-(5-chioiOpyridin-2-yl)-5-oxo-7H-pyrroio[3,4-b]pyrazin-7-yTj 4-methylpiperazine-l -
carboxylate
Figure imgf000020_0001
pharmaceutically acceptable salt thereof.
[0137] In some embodiments,“suvorexant” may also be referred to as Belsomra®, [(7R)-4-(5- chloro~l,3-benzoxazol-2-yl)~7-methyl~l,4-diazepan-i-yl]-[5-methyl~2-(tnazol-2-
yl)phenyl]methanone
Figure imgf000020_0002
pharmaceutically acceptable salt thereof.
Methods of Use
[0138] Sleep fragmentation, a condition in humans characterized by poor sleep consolidation, frequent brief arousals or microarousals (defined by the American Academy of Sleep Medicine as episodes of cortical EEG activation lasting at least 2 seconds and up to 16 seconds in duration and interrupting sleep), and frequent transitions to lighter stages of sleep, results in significant daytime impairment that may include impaired attention and concentration, excessive sleepiness, impaired j udgement, impaired memory and learning, increased risk of accidents, and secondary morbidity and mortality when sleep fragmentation is a concomitant of pain, sleep disordered breathing, and other disease states. Patients suffering from sleep fragmentation are often unaware of the hundreds of brief arousals that may occur during the night, and primarily complain of severe daytime impairment. Sleep fragmentation patients often complain that their sleep is not beneficial, refreshing, or restorative.
[0139] The diagnosis of sleep fragmentation and its methods of treatment must not be equated with insomnia, which is a separate and distinct medical diagnosis. Insomnia is typically characterized by patient awareness and dissatisfaction with their sleep. Most insomnia patients have a hyperarousal disorder that makes it difficult to fall asleep and/or difficult to stay asleep but enable them to function well during the daytime. Insomnia is diagnosed by measuring the latency to persistent sleep, i.e., LPS (LPS of >30 minutes satisfies the definition of sleep-onset insomnia) and/or measuring the amount of time awake after sleep onset, i.e., WASQ (WASO of >50 minutes satisfies the definition of sleep-maintenance insomnia). It is common for insomnia patients, and particularly the elderly, to awaken in the middle of the night and be unable to return to sleep. Unlike patients suffering from sleep fragmentation, insomnia patients are almost always highly aware of their inability to fall asleep or stay asleep at night and complain about their nighttime experience, and typically do not complain about daytime impairment.
[0140] The present disclosure provides for the use of a preclinical sleep- wake and physiological assay system, named SCORE-2000® , or“SCORE™”. The SCORE™ pharmacology database includes standardized sleep-wake, physiological, and behavioral data for over 500 distinct compounds. The standardized nature of the experimental designs, data quality' control, and data analysis methods enable qualitative and quantitative comparisons between compounds.
[0141] The present disclosure pertains to the identification of pharmacological compounds that are useful for treating sleep disorders characterized in whole or m part by sleep fragmentation.
An extensive body of research heretofore undisclosed has identified compounds that can reduce sleep fragmentation by (i) reducing the number of arousal s (measured preclinically as reduced number of wake bouts per hour) and by (ti) increasing sleep consolidation (measured
preclinically by average sleep bout duration per hour).
[0142] The utility of compounds of the present disclosure to improve sleep fragmentation was assessed by evaluating the sleep architecture and sleep quality endpoints of several hundred compounds with established soporific activity.
Rat and human sleep
[0143] The present disclosure provides a pre-clinica! drug evaluation using rats. Without washing to be bound by theory, rat sleep and human sleep have all of the necessary fundamental similarities to permit the rat to be used as a preclinical model.
[0144] As such, compounds that are soporific in a rat may have soporific effects in a human, and compounds that are soporific in a human may have soporific effects in a rat [0145] Both rat and human exhibit robust circadian modulation of sleep tendency and sleep architecture.
[0146] The“homeostatic” control of sleep shares similarity across mammalian species, including humans, in that loss of sleep increases a homeostatic drive for sleep evidenced by a reduction in latency to sleep onset, increase in the depth of sleep that can be reflected by the amount of low- frequency“delta” EEG (“EEG slow waves”) during nonREM, an increase in sleep consolidation as measured by sleep bout duration, or an increase in total sleep time. Sleep deprivation m a subject causes the subject to fall asleep faster, sleep deeper, sleep more efficiently (e.g., more consolidated bouts of sleep), and sleep more (e.g., an increase of sleep time) until the homeostatic drive for sleep becomes adequately discharged through the sleeping process.
[0147] Uninterrupted, well consolidated sleep can determine sleep quality in both a rat and a human. No matter how much a subject sleeps or what frequency of EEG dominates during sleep, the beneficial work of the sleeping process requires that sleep is not fragmented (interrupted) by frequent arousa!s.
[0148] Without wishing to be bound by theory, compounds of the present disclosure which affect REM sleep by decreasing the latency to sleep onset, increasing sleep time, increasing the depth and/or consolidation of sleep, reducing arousals, or a combination of the aforementioned effects in a subject, may have the same effects in a different subject. The compounds of the present disclosure, which affect REM sleep by decreasing the latency to sleep onset effects in a subject, may have the same effects on a subject of a different species. The compounds of the present disclosure, which affect REM sleep by decreasing the latency to sleep onset effects in rats, may have the same effects on a subject of different species. The compounds of the present disclosure, which affect REM sleep by decreasing the latency to sleep onset effects in rats, may have the same effects on a human.
[0149] Without washing to be bound by theory, compounds of the present disclosure which affect REM sleep by increasing sleep time m a subject, have the same effects in a different subject. The compounds of the present disclosure winch affect REM sleep by increasing sleep tune m a subject, may have the same effects on a subject of a different species. The compounds of the present disclosure which affect REM sleep by increasing sleep time in rats, may have the same effects on a subject of a different species. The compounds of the present disclosure which affect REM sleep by increasing sleep time in rats, may have the same effects on a human. [0150] Without wishing to he bound by theory, compounds of the present disclosure winch affect REM sleep by increasing the depth and/or consolidation of sleep m a subject, have the same effects in a different subject. Compounds of the present disclosure which affect REM sleep by increasing the depth and/or consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth and/or consolidation of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth and/or consolidation of sleep in rats, may have the same effects m a human.
[0151] Without wishing to be bound by theoiy, compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth and consolidation of sleep in rats, may have the same effects in a human.
[0152] Without wishing to be bound by theory, compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth or consolidation of sleep in rats, may have the same effects in a human.
[0153] Without wishing to be bound by theory, compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in a subject, may have the same effects in a subject of a different species.
Compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the depth of sleep in rats, may have the same effects in a human.
[0154] Without wishing to be bound by theory, compounds of the present disclosure winch affect REM sleep by increasing the consolidation of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect REM sleep by increasing the consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the consolidation of sleep m rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by increasing the consolidation of sleep m rats, may have the same effects in a human.
[0155] Without wishing to be bound by theoiy, compounds of the present disclosure winch affect REM sleep by reducing arousals in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect REM sleep by reducing arousals in a subject, have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by reducing arousals in rats, have the same effects in a subject of a different species. Compounds of the present disclosure which affect REM sleep by reducing arousals in rats, have the same effects in a human.
[0156] Without wishing to be bound by theory', compounds of the present disclosure which affect NREM sleep by decreasing the latency to sleep onset, increasing sleep time, increasing the depth and/or consolidation of sleep, reducing arousals, or a combination of the aforementioned effects in a subject, may have the same effects in a different subject. The compounds of the present disclosure, which affect NREM sleep by decreasing the latency to sleep onset effects in a subject, may have the same effects on a subject of a different species. The compounds of the present disclosure, which affect NREM sleep by decreasing the latency to sleep onset effects in rats, may have the same effects on a subject of different species. The compounds of the present disclosure, which affect NREM sleep by decreasing the latency to sleep onset effects in rats, may have the same effects on a human.
[0157] Without wishing to be bound by theory, compounds of the present disclosure which affect NREM sleep by increasing sleep time in a subject, have the same effects in a different subject. The compounds of the present disclosure which affect NREM sleep by increasing sleep time in a subject, may have the same effects on a subject of a different species. The compounds of the present disclosure which affect NREM sleep by increasing sleep time in rats, may have the same effects on a subject of a different species. The compounds of the present disclosure which affect NREM sleep by increasing sleep time in rats, may have the same effects on a human.
[0158] Without wishing to be bound by theory, compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep m a subject, have the same effects in a different subject. Compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth and/or consolidation of sleep in rats, may have the same effects in a human.
[0159] Without wishing to be bound by theory , compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep m a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth and consolidation of sleep in rats, may have the same effects in a human
[0160] Without wishing to be bound by theory , compounds of the present disclosure which affect NREM sleep by increasing the depth or consolidation of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect NREM sleep by- increasing the depth or consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth or consolidation of sleep in rats, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth or consolidation of sleep in rats, may have the same effects in a human.
[0161] Without washing to be bound by theory, compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in rats, may have the same effects m a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the depth of sleep in rats, may have the same effects in a human.
[0162] Without washing to be bound by theory, compounds of the present disclosure which affect NREM sleep by increasing the consolidation of sleep in a subject, have the same effects in a different subject. Compounds of the present disclosure winch affect NREM sleep by increasing the consolidation of sleep in a subject, may have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by increasing the consolidation of sleep in rats, may have the same effects m a subject of a different species.
Compounds of the present disclosure wiiich affect NREM sleep by increasing the consolidation of sleep in rats, may have the same effects in a human.
[0163] Without wishing to be bound by theory', compounds of the present disclosure which affect NREM sleep by reducing arousals in a subject, have the same effects in a different subject. Compounds of the present disclosure which affect NREM sleep by reducing arousals in a subject, have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by reducing arousals in rats, have the same effects in a subject of a different species. Compounds of the present disclosure which affect NREM sleep by reducing arousals in rats, have the same effects in a human.
[0164] Without wishing to be bound by theory, sleep continuity can be measured as the duration of NREM“bouts” or the duration of REM bouts, or the duration NREM+REM“bouts”, wherein an arousal or bout of wakefulness interrupts the NREM-REM cycle.
[ 0165] In some embodiments, sleep bout can be comprised of NREM, REM, or NREM+REM.
[0166] In some embodiments, sleep bout can be comprised of NREM. In some embodiments, sleep bout can be comprised of REM. In some embodiments, sleep bout can be comprised of
NREM+REM.
[ 0167] In some embodiments, NREM and REM sleep alternate in what may be called the NREM-REM cycle. In some embodiments NREM precedes REM.
[0168] In some embodiments, the proportion of time spent in NREM versus REM is the same for different subjects. In some embodiments, the proportion of time spent in NREM versus REM is the same for different subjects of different species. In some embodiments, the proportion of time spent in NREM versus REM is the same for a rat and a subject of a different species. In some embodiments, the proportion of time spent in NREM versus REM is the same for a rat and a human.
[0169] In some embodiments, the proportion of time spent in NREM versus REM is about 5: 1.
In some embodiments, the proportion of time spent m NREM versus REM is about 4: 1. In some embodiments, the proportion of time spent in NREM versus REM is about 3: 1. In some embodiments, the proportion of time spent in NREM versus REM is about 2: 1.
[0170] In some embodiments, the proportion of time spent in NREM versus REM is from about 100: 1 to about 1 : 1. In some embodiments, the proportion of time spent m NREM versus REM is from about 90: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 80: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 70: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 60: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 50: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 40: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 30: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 20: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 10: 1 to about 1 : 1. In some embodiments, the proportion of time spent in NREM versus REM is from about 5: 1 to about 1 : 1. In some embodiments, the species is a mouse. In some embodiments, the species is a hoofed animal. In some embodiments, the hoofed animal is a horse or cow. In some embodiments, the species is not a laboratory rat. In some embodiments, the species is not a human.
[0171] In some embodiments, hypnotics reduce REM sleep to some degree, and several classes of sleep disorder medicines can strongly suppress REM sleep. Without wishing to he bound by theory, REM sleep suppression is relevant to learning, memory, and/or psychiatric health.
[0172] Without wishing to he bound by theory, the relative effect of some classes of medicines for sleep disorders, neuropsychiatric disorders, and cardiovascular disease that either inhibit or stimulate REM sleep translates from a subject to a different subject. The relative effect of some classes of medicines for sleep disorders, neuropsychiatric disorders, and cardiovascular disease that either inhibit or stimulate REM sleep translates from a subject to a subject of a different species. The relative effect of some classes of medicines for sleep disorders, neuropsychiatric disorders, and cardiovascular disease that either inhibit or stimulate REM sleep translates from a rat to a subject of a different species. The relative effect of some classes of medicines for sleep disorders that either inhibit or stimulate REM sleep translate from rat to a human.
[0173] In some embodiments, a class of medicine which may inhibit or stimulate REM or NREM sleep is selected from a sleep therapeutic, a neuropsychiatric antidepressant, and a cardiovascular medicine.
[0174] In some embodiments, a sleep therapeutic is a sedative hypnotic GABAA positive allosteric modulators that binds to the GABAA benzodiazepine receptor. In some embodiments, a neuropsychiatric antidepressant is a selective serotonin reuptake inhibitor (SSRI) or an atypical antipsychotics. In some embodiments, a cardiovascular medicine is a therapeutic that may bind to an alpha-adrenergic receptor.
[0175] There are two differences which may be present between rat and human sleep. First, rats are night-active, whereas humans are day-active. This difference may have no importance per se for testing drug effects on sleep and wakefulness. The timing of the dose relative to the normal sleep period can be relied upon when evaluating drug efficacy on sleep related variables (e.g., inhibition of REM sleep) when comparing rat and human sleep. The difference between rats and humans is sleep-bout length, also referred to as“sleep continuity” Further, humans may consolidate sleep into a single period per day, interrupted normally only by short (e.g., less than 2 hours, less than 1 hour, less than 45 minutes, less than 30 minutes, less than 25 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 5 minutes, or less than 1 minute) bouts of wakefulness. The abnormal conditions may result in human sleep becoming highly fragmented, diminishing the restorative benefits of sleep. Rats may have shorter bouts of sleep that occur throughout the 24-hour day (e.g., on average, every 20 minutes, a rat completes a sleep-wake cycle). During darkness (when the rat may be most active), sleep typically occupies about 1/3 of each 20-minute cycle, and REM sleep is rare. During the day (lights-on), the rat typically sleeps about 2/3 of each 20-minute cycle. The polyphasic nature of sleep and shorter spontaneous sleep bout durations m the rat, enables highly sensitive assessments of drug effects, such as those that increase sleep consolidation (sleep bout duration), decrease the number of arousals (number of wake bouts), and a variety of secondary but desirable measures of sleep quality, for example, EEG slow wave activity in noriREM sleep, and measures of wake maintenance as measured by wake bout duration. Sleep bout-length may also be a sensitive measure of physiological sleepiness and is a pre-clinical predictor of soporific efficacy in humans.
Timing of treatment.
[0176] Empirical optimization can be performed by assessing sleep-related compounds by administering such compounds at two circadian times of day, CT-18 and CT-5, wherein CT-0 is defined as lights-on. CT-18 is the mid-point of the activity phase of the rat’s circadian cycle, 6 hours after lights-off, and may be especially sensitive to soporific drug effects on sleep bout length, although such effects can be observed at both CT-18 and CT-5. CT-5 begins several hours of peak abundance of REM sleep and thus is a sensitive time to reveal drug-related inhibition of REM sleep. Both CT-18 and CT-5 are suitable times of the day for the assessment of drug effects on sleep fragmentation as measured by arousals (number of wake bouts), sleep consolidation (sleep bout duration), as well as assessments of maintenance of wakefulness (wake bout length) and drug-related side effects.
[0177] Preclinica! effects observed at either CT-5 (treatments administered at a time of day corresponding to 5 hours after lights-on) and/or CT-18 (treatments administered at a time of day corresponding to 18 hours after lights-on or 6 hours after light-off when animals are housed in a 24 hour light dark cycle consisting of 12-hours of light and 12 hours of dark) are considered sufficient for purposes of identifying compounds which may reduce sleep fragmentation.
Biological Assay
[0178] The subject is surgically prepared for EEG and EMG recording and administered an analgesic with an antibiotic, followed by therapeutic deliver via intrapentoneal or oral administration. The sleep and wakefulness is determined using SCORE™.
[0179] Statistically significant differences between drug and vehicle may be screened using a post-hoc Student’s T-test applied to hourly binned data and adjusted for repeated measures.
[0180] The compounds of the present disclosure improve sleep fragmentation, as assessed by- evaluating the sleep architecture and sleep quality endpoints of several neurosteroid compounds with established affinity and functional activity at this target. [0181] In some embodiments, the sleep disorder is improved by (i) reducing the number of arousals fas measured by the number of wake bouts per hour), and (if) increasing sleep consolidation (as measured by average sleep bout duration per hour). In some embodiments, the sleep disorder is improved by (i) reducing the number of arousals (as measured by the number of wake bouts per hour), or (ii) increasing sleep consolidation (as measured by average sleep bout duration per hour). In some embodiments, the sleep disorder is improved by reducing the number of arousals (as measured by the number of wake bouts per hour. In some embodiments, sleep fragmentation is improved by increasing sleep consolidation (as measured by average sleep bout duration per hour).
Methods of Use
[0182] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of treating, or preventing a sleep disorder.
[0183] In some embodiments, the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold.
[0184] In some embodiments, the sleep disorder is increased disturbed sleep. In some embodiments, the sleep disorder is increased sleep fragmentation. In some embodiments, the sleep disorder is increased arousals. In some embodiments, the sleep disorder is decreased arousal threshold.
[0185] The present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds of the present disclosure or pharmaceutically acceptable salt thereof to a subject in need thereof.
[0186] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist, a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
[0187] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels or a2d-2 auxiliary subunit of voltage-gated calcium channels, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
[0188] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 and a2d- 2 auxiliary subunits of voltage-gated calcium channels, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
[0189] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels, or pharmaceutically acceptable salts thereof, to a subject in need thereof.
[0190] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, or pharmaceutically acceptable salts thereo to a subject in need thereof.
[0191] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a 5HT2A receptor antagonist and inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0192] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a 5HT2A receptor antagonist or a 5HT2A receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0193] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a 5HT2A receptor antagonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0194] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a SHTZA receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0195] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor antagonist and inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof. [0196] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor antagonist or a Hi receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0197] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor antagonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0198] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a Hi receptor inverse agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0199] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a ligand of a a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a a2d-2 auxiliary subunit of voltage-gated calcium channels, a gamma-hydroxy butyrate (GHB) receptor ligand, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0200] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a GABAB receptor agonist, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0201] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0202] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of or treating a sleep disorder by administering one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0203] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of a sleep disorder by administering one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxy butyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0204] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserm, and doxepin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
[0205] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of or treating a sleep disorder by administering one or more compounds selected from gabapentin, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a
pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0206] In some embodiments, the present disclosure is directed to a method of alleviating a symptom of a sleep disorder by administering one or more compounds selected from gabapentin, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0207] In some embodiments, the present disclosure provides one or more compounds selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0208] In some embodiments, the present disclosure provides one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use m alleviating a symptom of or treating a sleep disorder.
[0209] In some embodiments, the present disclosure provides one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use m alleviating a symptom of a sleep disorder.
[0210] In some embodiments, the present disclosure provides one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder. [021 1] In some embodiments, the present disclosure provides one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of or treating a sleep disorder.
[0212] In some embodiments, the present disclosure provides one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of a sleep disorder.
[0213] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, mirogabalm, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0214] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of or treating a sleep disorder.
[0215] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of a sleep disorder.
[0216] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0217] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of or treating a sleep disorder.
[0218] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentin, pregabalm, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, for alleviating a symptom of a sleep disorder. [0219] In some embodiments, the present disclosure provides the use of one or more compounds selected from gahapentin, pregabafm, mirogabalin, trazodone, nelotanserm, pimavanserm, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0220] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of or treating a sleep disorder.
[0221] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of a sleep disorder.
[0222] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserm, and doxepin, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0223] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserin, and doxepin, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of or treating a sleep disorder.
[0224] In some embodiments, the present disclosure provides the use of one or more compounds selected from gabapentm, pregabalin, trazodone, nelotanserin, pimavanserm, and doxepin, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of sleep a sleep disorder.
[0225] In some embodiments, the compound is gabapentm, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, or gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
[0226] In some embodiments, the compound is gabapentm, pregabalin, or mirogabalin, or a pharmaceutically acceptable salt thereof. [0227] In some embodiments, the compound is gabapentm or pregabalm, or a pharmaceutically acceptable salt thereof.
[0228] In some embodiments, the compound is trazodone, nelotanserm, or pimavanserin, or a pharmaceutically acceptable salt thereof.
[0229] In some embodiments, the compound is doxepin or diphenhydramine, or a
pharmaceutically acceptable salt thereof.
[0230] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering gabapentin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
[0231] In some embodiments, the present disclosure provides gabapentin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0232] In some embodiments, the present disclosure provides the use of gabapentin, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0233] In some embodiments, the present disclosure provides the use of gabapentin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0234] In some embodiments, the compound is gabapentm, or a pharmaceutically acceptable salt thereof.
[0235] In some embodiments, gabapentin is administered at a dose between about 0.010 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 0.05 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 0.1 mg/kg and about 500 mg/kg. In some embodiments, gabapentm is
administered at a dose between about 0.5 mg/kg and about 500 mg/kg. In some embodiments, gabapentm is administered at a dose between about 1 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 5 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 400 mg/kg. In some embodiments, gabapentm is administered at a dose between about 10 mg/kg and about 300 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 200 mg/kg. In some embodiments, gabapentin is administered at a dose between about 10 mg/kg and about 100 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 500 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 400 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 300 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 200 mg/kg. In some embodiments, gabapentin is administered at a dose between about 50 mg/kg and about 100 mg/kg. In some embodiments, gabapentin is
administered at a dose between about 75 mg/kg and about 300 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 200 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 150 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 125 mg/kg. In some embodiments, gabapentin is administered at a dose between about 75 mg/kg and about 100 mg/kg.
[0236] In some embodiments, gabapentin is administered at a dose of about 0.010 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.015 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.020 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.025 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.05 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.1 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.2 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.3 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.4 mg/kg. In some embodiments, gabapentin is administered at a dose of about 0.5 mg/kg. In some embodiments, gabapentin is administered at a dose of about 1 mg/kg. In some embodiments, gabapentin is administered at a dose of about 2 mg/kg. In some embodiments, gabapentin is administered at a dose of about 3 mg/kg. In some embodiments, gabapentin is administered at a dose of about 4 mg/kg. In some embodiments, gabapentin is administered at a dose of about 5 mg/kg. In some embodiments, gabapentin is administered at a dose of about 6 mg/kg. In some embodiments, gabapentin is administered at a dose of about 7 mg/kg. In some embodiments, gabapentin is administered at a dose of about 8 mg/kg. In some embodiments, gabapentin is administered at a dose of about 9 mg/kg. In some embodiments, gabapentin is administered at a dose of about 10 mg/kg. In some embodiments, gabapentin is administered at a dose of about 15 mg/kg. In some embodiments, gabapentin is administered at a dose of about 20 mg/kg. In some embodiments, gabapentm is administered at a dose of about 25 mg/kg. In some embodiments, gabapentin is administered at a dose of about 50 mg/kg. In some embodiments, gabapentin is administered at a dose of about 75 mg/kg. In some embodiments, gabapentm is administered at a dose of about 100 mg/kg. In some embodiments, gabapentin is administered at a dose of about 125 mg/kg. In some embodiments, gabapentin is administered at a dose of about 150 mg/kg. In some embodiments, gabapentin is administered at a dose of about 175 mg/kg. In some embodiments, gabapentin is administered at a dose of about 200 mg/kg.
[0237] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering pregabalin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
[0238] In some embodiments, the present disclosure provides pregabalin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0239] In some embodiments, the present disclosure provides the use of pregabalin, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0240] In some embodiments, the present disclosure provides the use of pregabalin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0241 ] In some embodiments, the compound is pregabalin, or a pharmaceutically acceptable salt thereof.
[0242] In some embodiments, pregabalin is administered at a dose between about 0.010 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 0.05 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 0.1 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 0.5 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 45 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 35 mg/kg. In some embodiments, pregabalin is administered at a dose between 1 mg/kg and 30 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 25 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 20 mg/kg. In some embodiments, pregabalin is administered at a dose between about 1 mg/kg and about 15 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 25 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 20 mg/kg. In some embodiments, pregabalin is administered at a dose between about 5 mg/kg and about 15 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 30 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 25 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 20 mg/kg. In some embodiments, pregabalin is administered at a dose between about 10 mg/kg and about 15 mg/kg.
[0243] In some embodiments, pregabalin is administered at a dose of about 0.010 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.015 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.020 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.025 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.05 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.1 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.2 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.3 mg/kg. In some embodiments, pregabalin is administered at a dose of about 0.4 mg/kg. In some
embodiments, pregabalin is administered at a dose of about 0.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 1 mg/kg. In some embodiments, pregabalin is administered at a dose of about 1.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 2 mg/kg. In some embodiments, pregabalin is administered at a dose of about 2.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 3 mg/kg. In some embodiments, pregabalin is administered at a dose of about 3.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 4 mg/kg. In some embodiments, pregabalin is administered at a dose of about 4.5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 5 mg/kg. In some embodiments, pregabalin is administered at a dose of about 10 mg/kg. In some embodiments, pregabalin is administered at a dose of about 15 mg/kg. In some embodiments, pregabalin is administered at a dose of about 20 mg/kg. In some embodiments, pregabalm is administered at a dose of about 25 mg/kg. In some embodiments, pregabalin is administered at a dose of about 30 mg/kg. In some embodiments, pregabalin is administered at a dose of about 35 mg/kg. In some embodiments, pregabalin is administered at a dose of about 40 mg/kg. In some embodiments, pregabalin is administered at a dose of about 45 mg/kg. In some embodiments, pregabalin is administered at a dose of about 50 mg/kg.
[0244] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering mirogabalin, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
[0245] In some embodiments, the present disclosure provides mirogabalin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0246] In some embodiments, the present disclosure provides the use of mirogabalin, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0247] In some embodiments, the present disclosure provides the use of mirogabalin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0248] In some embodiments, the compound is mirogabalin, or a pharmaceutically acceptable salt thereof.
[0249] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering trazodone, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0250] In some embodiments, the present disclosure provides trazodone, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0251] In some embodiments, the present disclosure provides the use of trazodone, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0252] In some embodiments, the present disclosure provides the use of trazodone, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder. [0253] In some embodiments, the compound is trazodone, or a pharmaceutically acceptable salt thereof.
[0254] In some embodiments, trazodone is administered at a dose between about 0.010 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 0.05 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 0.1 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 0.5 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 90 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 80 mg/kg.
In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 70 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 60 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 1 mg/kg and about 30 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 100 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 90 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 80 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 70 mg/kg.
In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 60 rng/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 50 rng/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 60 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 10 mg/kg and about 30 g/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 60 g/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 15 mg/kg and about 30 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 60 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 50 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 40 mg/kg. In some embodiments, trazodone is administered at a dose between about 20 mg/kg and about 30 mg/kg.
[0255] In some embodiments, trazodone is administered at a dose of about 0.010 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.015 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.020 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.025 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.05 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.1 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.2 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.3 mg/kg. In some
embodiments, trazodone is administered at a dose of about 0.4 mg/kg. In some embodiments, trazodone is administered at a dose of about 0.5 mg/kg. In some embodiments, trazodone is administered at a dose of about 1 mg/kg. In some embodiments, trazodone is administered at a dose of about 2 mg/kg. In some embodiments, trazodone is administered at a dose of about 3 mg/kg. In some embodiments, trazodone is administered at a dose of about 4 mg/kg. In some embodiments, trazodone is administered at a dose of about 5 mg/kg. In some embodiments, trazodone is administered at a dose of about 10 mg/kg. In some embodiments, trazodone is administered at a dose of about 15 mg/kg. In some embodiments, trazodone is administered at a dose of about 20 mg/kg. In some embodiments, trazodone is administered at a dose of about 25 mg/kg. In some embodiments, trazodone is administered at a dose of about 30 mg/kg. In some embodiments, trazodone is administered at a dose of about 35 mg kg. In some embodiments, trazodone is administered at a dose of about 40 mg/kg. In some embodiments, trazodone is administered at a dose of about 45 mg/kg. In some embodiments, trazodone is administered at a dose of about 50 mg/kg.
[0256] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering nelotanserin, or a pharmaceutically acceptable salt thereof, to a subject in need thereof. [0257] In some embodiments, the present disclosure provides nelotansenn, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0258] In some embodiments, the present disclosure provides the use of nefotanserin, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0259] In some embodiments, the present disclosure provides the use of nelotanserin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0260] In some embodiments, the compound is nelotanserin, or a pharmaceutically acceptable salt thereof.
[0261] In some embodiments, nelotanserin is administered at a dose between about 0.010 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 0.05 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 0.1 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is
administered at a dose between about 0.5 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 45 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 35 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 25 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 20 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 1 mg/kg and about 15 mg/kg. In some embodiments, nelotansenn is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 5 mg/kg and about 25 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 5 mg/kg and about 20 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 5 mg/kg and about 15 mg/kg. In some embodiments, nelotansenn is administered at a dose between about 10 mg/kg and about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose between about 10 nig/kg and about 25 nig/kg. In some embodiments, nelotanserin is administered at a dose between about 10 mg/kg and about 20 mg/kg. In some embodiments, nelotansenn is administered at a dose between about 10 mg/kg and about 15 mg/kg.
[0262] In some embodiments, nelotanserin is administered at a dose of about 0.010 g kg. In some embodiments, nelotanserin is administered at a dose of about 0.015 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.020 mg/kg. In some
embodiments, nelotanserin is administered at a dose of about 0.025 mg/kg. In some
embodiments, nelotanserin is administered at a dose of about 0.05 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.1 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.2 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.3 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.4 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 0.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 1 mg/kg. In some
embodiments, nelotanserin is administered at a dose of about 1.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 2 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 2.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 3 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 3.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 4 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 4.5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 5 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 10 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 15 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 20 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 25 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 30 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 35 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 40 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 45 mg/kg. In some embodiments, nelotanserin is administered at a dose of about 50 mg/kg.
[0263] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering pimavanserin, or a pharmaceutically acceptable salt thereof, to a subject in need thereof [0264] In some embodiments, the present disclosure provides pimavanserm, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0265 ] In some embodiments, the present disclosure provides the use of pimavanserin, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0266] In some embodiments, the present disclosure provides the use of pimavanserin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0267] In some embodiments, the compound is pimavanserin, or a pharmaceutically acceptable salt thereof.
[0268] In some embodiments, pimavanserin is administered at a dose between about 0.010 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 0.05 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 0.1 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 0.5 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 500 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 400 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 300 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 1 mg/kg and about 200 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 1 mg/kg and about 100 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 1 mg/kg and about 60 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 10 mg/kg and about 500 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 10 mg/kg and about 400 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 10 mg/kg and about 300 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 10 mg/kg and about 200 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 10 mg/kg and about 100 mg/kg. In some embodiments, pimavanserm is administered at a dose between about 10 mg/kg and about 60 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 30 mg/kg and about 100 mg/kg. In some embodiments, pimavanserin is administered at a dose between about 30 mg/kg and about 60 mg/kg.
[0269] In some embodiments, pimavanserin is administered at a dose of about 0.010 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.01 5 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.020 mg/kg. In some
embodiments, pimavanserin is administered at a dose of about 0.025 mg/kg. In some
embodiments, pimavanserin is administered at a dose of about 0.05 mg/kg. In some
embodiments, pimavanserin is administered at a dose of about 0.1 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.2 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.3 mg/kg. In some embodiments, pimavanserin is
administered at a dose of about 0.4 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 0.5 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 1 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 2 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 3 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 4 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 5 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 6 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 7 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 8 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 9 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 10 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 25 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 30 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 50 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 60 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 75 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 100 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 125 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 150 mg/kg. In some embodiments, pimavanserin is administered at a dose of about 175 mg/kg. In some
embodiments, pimavanserin is administered at a dose of about 200 mg/kg. [0270] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder by administering doxepm, or a pharmaceutically acceptable salt thereof, to a subject m need thereof.
[0271] In some embodiments, the present disclosure provides doxepin, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0272] In some embodiments, the present disclosure provides the use of doxepin, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0273] In some embodiments, the present disclosure provides the use of doxepin, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0274] In some embodiments, the compound is doxepin, or a pharmaceutically acceptable salt thereof.
[0275] In some embodiments, doxepin is administered at a dose between about 0.010 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 0.05 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 0.1 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 0.5 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 50 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 45 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 40 mg/kg. In some embodiments, doxepm is administered at a dose between about 1 mg/kg and about 35 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 30 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 25 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 20 mg/kg. In some embodiments, doxepin is administered at a dose between about 1 mg/kg and about 15 mg/kg. In some embodiments, doxepin is administered at a dose between about 5 mg/kg and about 30 mg/kg. In some embodiments, doxepm is administered at a dose between about 5 mg/kg and about 25 mg/kg. In some embodiments, doxepm is
administered at a dose between about 5 mg/kg and about 20 mg/kg. In some embodiments, doxep is administered at a dose between about 5 mg/kg and about 15 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 30 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 25 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 20 mg/kg. In some embodiments, doxepin is administered at a dose between about 10 mg/kg and about 15 mg/kg.
[0276] In some embodiments, doxepin is administered at a dose of about 0.010 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.015 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.020 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.025 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.05 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.1 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.2 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.3 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.4 mg/kg. In some embodiments, doxepin is administered at a dose of about 0.5 mg/kg. In some embodiments, doxepin is administered at a dose of about 1 mg/kg. In some embodiments, doxepin is administered at a dose of about 2 mg/kg. In some embodiments, doxepin is administered at a dose of about 3 mg/kg. In some embodiments, doxepin is administered at a dose of about 4 mg/kg. In some embodiments, doxepin is administered at a dose of about 5 mg/kg. In some embodiments, doxepin is administered at a dose of about 10 mg/kg. In some embodiments, doxepin is administered at a dose of about 15 mg/kg. In some embodiments, doxepin is administered at a dose of about 20 mg/kg. In some embodiments, doxepin is administered at a dose of about 25 mg/kg. In some embodiments, doxepin is administered at a dose of about 30 mg/kg. In some embodiments, doxepin is administered at a dose of about 35 mg/kg. In some embodiments, doxepin is administered at a dose of about 40 mg/kg. In some embodiments, doxepin is administered at a dose of about 45 mg/kg. In some embodiments, doxepin is administered at a dose of about 50 mg/kg.
[0277] In some embodiments, the compound is administered orally. In some embodiments, the compound is administered parenteralfy. In some embodiments, the compound is administered transderma!ly. In some embodiments, the compound is administered subcutaneously. In some embodiments, the compound is administered intravenously. [0278] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering diphenhydramine, or a
pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0279] In some embodiments, the present disclosure provides diphenhydramine, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0280] In some embodiments, the present disclosure provides the use of diphenhydramine, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0281] In some embodiments, the present disclosure provides the use of diphenhydramine, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0282] In some embodiments, the compound is diphenhydramine, or a pharmaceutically acceptable salt thereof.
[0283] In some embodiments, the present disclosure provides a method of alleviating a symptom of, treating, or preventing a sleep disorder, by administering gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
[0284] In some embodiments, the present disclosure provides gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder.
[0285] In some embodiments, the present disclosure provides the use of gamma- hydroxybutyrate, for alleviating a symptom of, treating, or preventing a sleep disorder
[0286] In some embodiments, the present disclosure provides the use of gamma- hydroxybutyrate, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a sleep disorder.
[0287] In some embodiments, the compound is gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof
[0288] In some embodiments, the compound of the present disclosure is not suvorexant, zolpidenr, S-zopiclone, or a pharmaceutically acceptable salt thereof.
[0289] In some embodiments, the compound of the present disclosure is not suvorexant, or a pharmaceutically acceptable salt thereof. [0290] In some embodiments, the compound of the present disclosure is not zolpidem, or a pharmaceutically acceptable salt thereof.
[0291] In some embodiments, the compound of the present disclosure is not S-zopiclone, or a pharmaceutically acceptable salt thereof.
[0292] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respirator disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
[0293] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by sleep apnea, restless legs syndrome, a high respirator}' disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
[0294] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with sleep apnea, restless legs syndrome, high respiratory disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
[0295] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with sleep apnea.
[0296] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by sleep apnea.
[0297] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with sleep apnea.
[0298] In some embodiments, sleep apnea is obstructive sleep apnea.
[0299] In some embodiments, sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea.
[0300] In some embodiments, sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory' event related arousal (RERA) with a concomitant apnea.
[0301] In some embodiments, sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant hypopnea.
[0302] In some embodiments, sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) wi th a concomitant hypopnea.
[0303] In some embodiments, sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without concomitant acute hemoglobin desaturation.
[0304] In some embodiments, sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with concomitant acute hemoglobin desaturation.
[0305] In some embodiments, sleep apnea is central sleep apnea.
[0306] In some embodiments, sleep apnea is low-arousal threshold sleep apnea. [0307] In some embodiments, sleep apnea is hypopnea.
[0308] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with restless legs syndrome.
[0309] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by restless legs syndrome.
[0310] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with restless legs syndrome.
[031 1] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a high respiratory disturbance index
(RDI).
[0312] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a high respiratory disturbance index (RDI).
[0313] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a high respiratory disturbance index (RDI).
[0314] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea.
[0315] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with a concomitant apnea. [0316] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with or without a concomitant hypopnea.
[0317] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with a concomitant hypopnea.
[0318] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with or without concomitant acute hemoglobin desaturation.
[0319] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with concomitant acute hemoglobin desaturation.
[0320] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with or without concomitant hemoglobin desaturation.
[0321] In some embodiments, the RDI is associated with an elevated respiratory event related arousal (RERA) with concomitant hemoglobin desaturation.
[0322] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a neurological disease.
[0323] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a neurological disease.
[0324] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a neurological disease.
[0325] In some embodiments, the neurological disease is a neurodegenerative disease.
[0326] In some embodiments, the neurodegenerative disease is Lewy body disease (i.e., Lewy body dementia). In some embodiments, the Lewy body disease is diffuse.
[0327] In some embodiments, the neurodegenerative disease is amyotrophic lateral sclerosis (ALS). In some embodiments, the neurodegenerative disease is Huntington’s disease. In some embodiments, the neurodegenerative disease is Parkinson’s disease. In some embodiments, the neurodegenerative disease is Alzheimer’s disease. In some embodiments, the neurodegenerative disease is a synucleinopathy.
[0328] In some embodiments, a synucleinopathy is Alzheimer’s disease, Parkinson’s disease, or Lewy body dementia. In some embodiments, a synucleinopathy is Alzheimer’s disease. In some embodiments, a synucleinopathy is Parkinson’s disease. In some embodiments, a synucleinopathy is dementia with Lewy bodies. In some embodiments, a synucleinopathy is multiple system atrophy.
[0329] In some embodiments, the neurological disease is a neurodevelopmental disease. In some embodiments, the neurodevelopmental disease is autism. In some embodiments, the neurological disease is a muscular dystonia. In some embodiments, the dystonia is neuromuscular dystonia. In some embodiments, the neuromuscular dystonia is spasmodic torticollis.
[0330] In some embodiments, the neurological disease is multiple sclerosis (MS).
[0331] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a circadian rhythm disorder.
[0332] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a circadian rhythm disorder.
[0333] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a circadian rhythm disorder.
[0334] In some embodiments, the circadian rhythm disorder is advanced sleep- wake phase disorder. In some embodiments, the circadian rhythm disorder is irregular sleep-wake rhythm disorder. In some embodiments, the circadian rhythm disorder is jet lag. In some embodiments, the circadian rhythm disorder is shift work sleep disorder. In some embodiments, the circadian rhythm disorder is delayed sleep phase syndrome. In some embodiments, the circadian rhythm disorder is non-24 hour rhythm disorder. [0335] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with pain.
[0336] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by pain.
[0337] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with pain.
[0338] In some embodiments, the pain is selected from an inflammatory' pain, a nociceptive pain, a neuropathic pain, a mixed nociceptive and neuropathic pain, a post-operative pain, a post herpetic pain, a traumatic pain, a phantom-limb pain, a fibromyalgia, a back pain, a cancer pain, and an osteoart.hr itic pain.
[0339] In some embodiments, the pain is an inflammatory pain. In some embodiments, the inflammatory pain is arthritis. In some embodiments, the arthritis is rheumatoid arthritis. In some embodiments, the arthritis is osteoarthritis.
[0340] In some embodiments, the pain is a nociceptive pain. In some embodiments, the nociceptive pain is acute. In some embodiments, the nociceptive pain is chronic. In some embodiments, the nociceptive pain is caused by a cancer therapy. In some embodiments, the nociceptive pain is caused by a surgery.
[0341] In some embodiments, the pain is a neuropathic pain. In some embodiments, the neuropathic pam is chronic. In some embodiments, the neuropathic pain is acute. In some embodiments, the neuropathic pam is back pam. In some embodiments, the neuropathic pam is caused by a spinal cord injury. In some embodiments, the neuropathic pain is caused by multiple sclerosis. In some embodiments, the neuropathic pam is caused by a stroke. In some
embodiments, the neuropathic pain is caused by diabetes. In some embodiments, the neuropathic pain is caused by a metabolic condition.
[0342] In some embodiments, the pain is a mixed nociceptive and neuropathic pain. [0343] In some embodiments, the pain is a post-operative pain.
[0344] In some embodiments, the pain is a post-herpetic pain.
[0345] In some embodiments, the pain is a traumatic pain. In some embodiments, traumatic pain is caused by causalgia.
[0346] In some embodiments, the pam is a phantom-limb pain.
[0347] In some embodiments, the pain is a fibromyalgia.
[0348] In some embodiments, the pam is a back pam. In some embodiments, the pam is a low back pain.
[0349] In some embodiments, the pam is a cancer pain. In some embodiments, the cancer pain is cancer. In some embodiments, the cancer pain is caused by a tumor. In some embodiments, the cancer pain is caused by a cancer treatment. In some embodiments, the cancer pain is caused by chemotherapy. In some embodiments, the cancer pain is radiation therapy. In some
embodiments, the cancer pam is caused by surgery.
[0350] In some embodiments, the pain is an osteoarthritic pain
[0351] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with periodic leg movement disorder
(PLMD).
[0352] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by periodic leg movement disorder (PLMD).
[0353] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with periodic leg movement disorder (PLMD).
[0354] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with REM behavior disorder. [0355] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by REM behavior disorder.
[0356] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with REM behavior disorder.
[0357] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with elderly fragmented sleep.
[0358] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by elderly fragmented sleep.
[0359] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with elderly fragmented sleep.
[0360] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with age-related sleep fragmentation.
[0361] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousais, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by age-related sleep fragmentation.
[0362] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with age-related sleep fragmentation.
[0363] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with post-menopausal sleep disorder.
[0364] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by post-menopausal sleep disorder.
[0365] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with post-menopausal sleep disorder.
[0366] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with substance abuse.
[0367] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by substance abuse.
[0368] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with substance abuse.
[0369] In some embodiments, the substance abuse is opioid abuse or alcoholism. In some embodiments, the substance abuse is opioid abuse. In some embodiments, the substance abuse is alcoholism.
[0370] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with substance abuse withdrawal.
[0371] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by substance abuse withdrawal.
[0372] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with substance abuse withdrawal.
[0373] In some embodiments, the substance abuse withdrawal is opioid withdrawal or alcohol withdrawal. In some embodiments, the substance abuse withdrawal is opioid withdrawal. In some embodiments, the substance abuse is alcohol withdrawal.
[0374] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with narcolepsy.
[0375] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject wherein the sleep disorder is caused by narcolepsy.
[0376] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with narcolepsy.
[0377] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with a mental disorder.
[0378] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by a mental disorder.
[0379] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with a mental disorder.
[0380] In some embodiments, the mental disorder is depression, major depressive disorder, post- traumatic stress disorder, anxiety disorder, bipolar disorder, or schizophrenia.
[0381] In some embodiments, the mental disorder is depression. In some embodiments, the mental disorder is major depressive disorder. In some embodiments, the mental disorder is post- traumatic stress disorder. In some embodiments, the mental disorder is anxiety disorder. In some embodiments, the mental disorder is bipolar disorder. In some embodiments, the mental disorder is schizophrenia.
[0382] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with non-restorative sleep.
[0383] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by non-restorative sleep.
[0384] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with non-restorative sleep.
[0385] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with snoring.
[0386] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousafs, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by snoring.
[0387] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with snoring.
[0388] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by or co-morbid with idiopathic hypersomnia.
[0389] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is caused by idiopathic hypersomnia.
[0390] In some embodiments, the present disclosure is directed to alleviating a symptom of, treating, or preventing a sleep disorder wherein the sleep disorder is increased disturbed sleep, increased sleep fragmentation, increased arousals, or decreased arousal threshold in a subject, wherein the sleep disorder is co-morbid with idiopathic hypersomnia.
[0391] In some embodiments, the number of arousals is decreased by up to about 99%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, about 50%, about 45%, about 40%, about 35%, about 30%, about 25%, about 20%, about 15%, about 10%, or about 5% reduction in the number of wake bouts per hour post-treatment relative to vehicle treatment.
[0392] In some embodiments, gabalin alleviates a symptom of, treats, or prevents a sleep disorder.
[0393] In some embodiments, gabapentin decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment. In some embodiments, gabapentin increases sleep consolidation as demonstrated by >2-foJd increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, gabapentin increases EEG slow' wave activity in nonREM sleep. [0394] In some embodiments, pregabalin alleviates a symptom of, treats, or prevents a sleep disorder.
[0395] In some embodiments, pregabalin decreases the number of arousals (approximately 40% reduction in the number of wake bouts per hour post-treatment relative to vehicle treatment). In some embodiments, pregabalin increases sleep consolidation as evidenced by a nearly 3 -fold increase in average sleep bout duration per hour post-treatment relative to vehicle treatment.
[0396] In some embodiments, mirogabaiin alleviates a symptom of, treats, or prevents a sleep disorder.
[0397] In some embodiments, mirogabaiin decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment. In some
embodiments, mirogabaiin increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, mirogabaiin increases EEG slow wave activity in nonREM sleep.
[0398] In some embodiments, trazodone alleviates a symptom of, treats, or prevents a sleep disorder.
[0399] In some embodiments, trazodone decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment. In some
embodiments, trazodone increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some
embodiments, trazodone increases EEG slow wave activity in nonREM sleep.
[0400] In some embodiments, nelotansenn alleviates a symptom of, treats, or prevents a sleep disorder.
[0401] In some embodiments, nelotansenn decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment. In some
embodiments, nelotansenn increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, neJotanserin increases EEG slow wave activity in nonREM sleep.
[0402] In some embodiments, pimavanserin alleviates a symptom of, treats, or prevents a sleep disorder.
[0403] In some embodiments, pimavanserin decreases arousals as demonstrated by a reduced number of w¾ke bouts per hour post-treatment relative to vehicle treatment. In some embodiments, pimavanserin increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, pimavanserin increases EEG slow wave activity in nonREM sleep.
[0404] In some embodiments, trazodone, nelotanserm, and pimavanserin reduce sleep fragmentation.
[0405] In some embodiments, trazodone decreases the number of arousals (measured by the number of wake bouts per hour post-treatment relative to vehicle treatment). In some embodiments, trazodone increases sleep consolidation.
[0406] In some embodiments, nelotanserin decreases the number of arousals (measured by the number of w¾ke bouts per hour post-treatment relative to vehicle treatment). In some embodiments, nelotanserin increases sleep consolidation (approximately 3 -fold increase m average sleep bout duration per hour post-treatment relative to vehicle treatment).
[0407] In some embodiments, pimavanserin decreases the number of arousals (measured by the number of wake bouts per hour post-treatment relative to vehicle treatment). In some embodiments, pimavanserin increases sleep consolidation (as measured by average sleep bout duration per hour post-treatment relative to vehicle treatment). The efficacy achieved with pimavanserin may be less than that observed for trazodone and nelotanserin. The comparatively smaller efficacy of pimavanserin on sleep consolidation (average sleep bout duration) may be due to an efficacy ceiling (maximum achievable magnitude of effect) as stepwise dose escalation up to 60 mg/kg produced no further increase in average sleep bout duration per hour.
[0408] In some embodiments, doxepin alleviates a symptom of, treats, or prevents a sleep disorder.
[0409] In some embodiments, doxepin reduces sleep fragmentation.
[0410] In some embodiments, doxepin decreases the number of arousals (measured by the number of w¾ke bouts per hour post-treatment relative to vehicle treatment). In some embodiments, doxepin increases sleep consolidation (approximately 2-fold increase in average sleep bout duration per hour post-treatment relative to vehicle treatment).
[041 1] In some embodiments, diphenydramine alleviates a symptom of, treats, or prevents a sleep disorder.
[0412] In some embodiments, diphenydramine decreases arousals as demonstrated by a reduced number of w¾ke bouts per hour post-treatment relative to vehicle treatment. In some embodiments, diphenydramine increases sleep consolidation as demonstrated by an increase in duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, diphenydramine increases EEG slow wave activity in nonREM sleep.
[0413] In some embodiments, gamma-hydroxybutyrate alleviates a symptom of, treats, or prevents a sleep disorder.
[0414] In some embodiments, gamma-hydroxybutyrate decreases arousals as demonstrated by a reduced number of wake bouts per hour post-treatment relative to vehicle treatment. In some embodiments, gamma-hydroxybutyrate increases sleep consolidation as demonstrated by an increase m duration of average sleep bouts per hour post-treatment relative to vehicle treatment. In some embodiments, gamma-hydroxybutyrate increases EEG slow7 wave activity7 in nonREM sleep.
[0415] In some embodiments, one or more compounds of the present disclosure produces improvements in sleep fragmentation in comparison to compounds prescribed comprising the pharmacological standard of care for insomnia. In some embodiments, the standard of care compounds for insomnia are, for example, but not limited to, the orexin antagonists (e.g., suvorexant (BELSOMRA®)) and non-benzodiazepine, benzodiazepine receptor dependent GABAA allosteric modulators (e.g., zolpidem (AMB1EN®) and eszopiclone (LUNESTA®)), which do not adequately improve the requisite precimical measures of sleep fragmentation.
[0416] In some embodiments, orexin OC1 /ΌC2 antagonist suvorexant fails to improve sleep fragmentation at 3 mg/kg, 10 mg/kg, and 30 mg/kg. In some embodiments, suvorexant does not reduce the number of arousals as measured by the number of wake bouts per hour, but instead, increases the number of arousals. In some embodiments, suvorexant produces small
improvements in sleep consolidation at the highest dose (30 mg/kg) as measured by average sleep bout duration per hour post-treatment.
[0417] In some embodiments, zolpidem, at doses sufficient to produce increases m soporific efficacy as measured by EEG slow wave activity (EEG delta power in nonREM sleep), fails to reduce the number of arousals as measured by the number of wake bouts per hour and fails to produce improvement in sleep consolidation as measured by average sleep bout duration per hour. Zolpidem can produce sedative effects (up to and including non-responsiveness of the subject); however, increasing the dose of zolpidem may be complicated by a short kinetic half- life and can be contraindicated due to significant unwanted side effects including, but not limited to, ITEM sleep inhibition, severe motor coordination impairment, memory impairment, rebound insomnia, and other unwanted adverse effects, or a combination thereof.
[0418] In some embodiments, S-zopiclone (marketed as LUNESTA®) fails to reduce the number of arousals as measured by the number of wake bouts per hour and produces only small improvements in sleep consolidation as measured by average sleep bout duration per hour. In some embodiments, the dose of S-zopiclone tested (i.e., from about 5 mg/kg to about 30 mg/kg) does not increase sleep consolidation above the levels exhibited during the baseline rest phase (lights-on; circadian time 0:00-12:00 prior to treatment). While the immediate sedative hypnotic effects of S-zopiclone can increase with dose, driving additional soporific efficacy with high doses is contraindicated due to dose-dependent unwanted side effects. In some embodiments, S- zopiclone decreases Locomotor Activity Intensity (LMAi). In some embodiments, S-zopiclone dose-dependently inhibits REM sleep, which may be important for memory and learning. In some embodiments, standard of care insomnia drugs (e.g., zolpidem and S-zopiclone) can also reduce muscle tone, which is not only important for ambulation (e.g., getting up from sleep to use the bathroom), but also may have unwanted consequences for subjects diagnosed with or at risk for obstructive sleep apnea. In some embodiments, relaxation of upper airway muscles is undesirable as it could worsen obstructive sleep apnea and its comorbid health risks. Direct electromyographic (EMG) assessment of muscle tone may reveal unwanted myorelaxation (decrease in skeletal muscle electrical activity) after treatment with S-zopiclone. The improvement in noriREM sleep time induced by S-zopiclone can be followed by an undesirable dose-dependent rebound interference in sleep that would not enable improvement in sleep fragmentation.
EXAMPLES
[0419] The following examples illustrate the disclosure. These examples are not intended to limit the scope of the present disclosure, but rather to provide guidance to the skilled artisan to prepare and use the methods of the present disclosure. While particular embodiments of the present disclosure are described, the skilled artisan will appreciate that various changes and modifications can be made without departing from the spirit and scope of the disclosure.
Abbreviations [ 0420] The following abbreviations are used in the examples, which follow'. This list is not meant to be an all-inclusive list of abbreviations used in the application as additional standard abbreviations, which are readily understood by those skilled in the art, can also be used in the examples.
C Celsius
EEG Electro-encephalograph
EMG electromyograph
g grams
HP CD (2-hydroxypropyl)- -cyclodextrin
Hz hertz
kg kilograms
mL milliliters
Example 1. Experimental details and methods for determining sleep continuity,
wakef lness, number of arousals, locomotor activity, and soporific efficacy
Animal preparation rats
[0421] Adult, male Wistar rats (approximately 270 g at time of surgery, Charles River
Laboratories) were anesthetized (2% isoflourane in 95/5 oxygen) and surgically prepared with a cranial implant that permitted chronic electro-encephalogram (LEG) and electromyogram (EMG) recording. Body temperature and locomotor activity were monitored via a miniature transmitter (Minimitter Series 4000 E-Mitter, Bend, OR) surgically placed in the abdomen during the same anesthetic event the cranial portion was implanted. The cranial implant consisted of stainless steel screws (2 frontal [+3.9 AP from bregma, ±2.0 ML] and 2 occipital [-6.4 AP, ±5.5 ML]) for EEG recording. Two Teflon-coated stainless steel wires were positioned under the nuchal trapezoid muscles for EMG recording. All leads were soldered to a miniature connector (Microtech, Boothwyn, PA) and gas sterilized with ethylene oxide prior to surgery. The implant assembly was affixed to the skull by the combination of the EEG recording screws,
cyanoacrylate applied between the hermetically sealed implant connector and skull, and dental acrylic. An analgesic (buprinorphine 0.3 mg/kg IP) was administered pre-operatively and daily SC for 2 days post-surgery. An antibiotic was administered before surgery (chloramphenicol 40 mg/kg IM) and for 7-10 days after surgery (Clavamox b.i.d). At least three weeks were allowed for surgical recovery prior to any data collection.
Recording environment [0422] Rats were housed individually within specially modified Nalgene® microisolator cages equipped with an ultra-low-torque slip-ring commutator and a custom polycarbonate filter-top riser. These cages were located within separate, ventilated compartments of a stainless steel sleep-wake recording chamber. Food and water were available ad libitum and the ambient temperature was 23±1° C. A 24-hr light-dark cycle (LD 12: 12) was maintained throughout the study using fluorescent light. Light intensity averaged 35-40 lux at mid-level inside the cage. Relative humidity averaged 50% approximately. Animals were undisturbed for two days before and after each treatment.
Automated data collection
[0423] Sleep and wakefulness were determined using SCORE™— a microcomputer-based sleep-wake and physiological monitoring system. Validation of the SCORE™ sleep stage identification algorithm m rodents and utility m pre-clinical drug evaluation have been previously described (Van Gelder etal. 1991 ; Edgar et al Psychopharmacology 1991, 105, 374; J Pharmacology & Experimental Therapeutics 1997, 283, 757; Seidel et al J Pharmacology & Experimental Therapeutics, 1995, 275, 263; ./. Pharmacology’ & Experimental Therapeutics, 1998, 285, 1073). For the studies described herein, the system monitored amplified EEG
(x 10,000, bandpass 1-30 Hz; initial digitization rate 400 Hz [Grass Corp., Quincy, MA]), integrated EMG (bandpass 10-100 Hz, RMS integration), and telemetered body temperature and non-specific locomotor activity (LMA), and drink- and food-related activity, from up to 150 rodents simultaneously. Arousal states were classified on-line as NREM sleep, REM sleep, wake, or theta-dominated wake every 10 seconds using EEG period and amplitude feature extraction and ranked membership algorithms. Individually taught EEG-arousal-state templates and EMG criteria differentiated states of arousal. LMA and drink-related activity' were automatically recorded as counts per minute, and body temperature was recorded each minute. LMA was detected in both horizontal and vertical planes by a customized telemetry' receiver (ER4000, Minimitter, Bend, OR) beneath the cage. Drink-related activity' and food-related activity' were detected by beam break sensors closely situated around recessed access portals to the lixit and the food bin, respectively. The beam break area for the food bin was, however, relatively large, and these data have not been validated as an endpoint for food consumpti on per se. Telemetry measures (LMA and body temperature) were not part of the SCORE arousal -state determination algorithm; thus, sleep-scoring and telemetry data were concurrent but independent measures. In addition to frequent on-tine inspection of the EEG and EMG signals, quality control of the data was assured by expert analysts with a minimum of 4 years of experience using a proprietary suite of programs (SCOREVIEW™ , Hypnion, Inc., Lexington, MA) that allowed data quality of all variables to be flexibly scrutinized at the level of (i) individual visual examination of raw EEG and EMG signals, (ii) individual hourly mean timeseries, and (Hi) group mean timesenes, using a combination of graphical and statistical assessments. An integrated relational database was updated with data quality control decisions for each individual treatment, and this database controlled ail subsequent use of these data. Complete, digitized raw EEG and integrated EMG data have been written to CD-R and are permanently archived for all treatments.
Treatments and drug preparation
[0424] Drug dose, route of administration, and timing of administration are described for each compound and variable within the data exemplifications. Where applicable, methy!ee!lulose vehicle was prepared as a sterile 0.25% solution of methylcellulose (15 centipoise, Sigma, St. Louis, MO., USA). HPitCD (American Maise Products, Indianapolis, IN) vehicle was initially prepared as a 50% stock solution (w/v in 0.9% NaCl) and then further diluted with 0.9% NaCl to achieve percent solutions as reported for respective studies. Intraperitoneal administration was delivered at a volume of 1 mL/kg Oral dosing was at a volume of 1 -2 mL/kg.
[0425] Drugs were weighed using a Metier AN205 analytical balance (d=0.01 mg). Compound was mixed with vehicle using a sterile 2 inL ground glass pestle and mortar until completely dissolved, and then transferred to a sterile Vacutainer (red top) tube. Solutions were agitated immediately before being drawn into a syringe. To administer the treatment, each rat was removed from its cage for about 60-90 seconds to be weighed and treated (the home cage is the recording cage in SCORE™ systems). Note that this procedure caused no prior sleep loss, unlike cases in which the animal must first be acclimated to a special recording chamber. Rats in this experiment lived permanently in their“home cage” within the recording chamber. Prior sleep loss— for instance, the“acclimation” commonly used by other investigators— significantly influences the measurement of sleep-wakefulness responses to drugs (Edgar et al
Psychopharmacology 1991 , 105, 374; Meltzer & Serpa, Drug Dev. Res., 1988, 14, 151).
Study design
[0426] The standard recording duration for SCORE data was not less than 30 hours before and after treatment. The 30 hours pre-treatment baseline recording was itself preceded by at least 24 hours in which the animal was undisturbed in the home/recording cage. Rats were randomly assigned to treatments in parallel groups. Some rats received more than one active treatment, in which cases at least 7 days“washout” elapsed between each treatment.
Statistical Analysis
[0427] Statistically significant differences between drug and vehicle were assessed using a post-hoc Student’s T-test applied to hourly binned data and adjusted for repeated measures. Figure formats and graphics conventions
[0428] Post-treatment detail plots the first five hours post- treatment. Variables were computed in 5-minute bins, aligned to the minute of treatment. The first time bin, labeled 0, represents the first 5 minutes post treatment.
[0429] Pre- and post-treatment time series plot ±30 hours before and after CT-18 treatments (or 29 hr before and 31 hr after CT-5 treatments). Treatment occurred at the beginning of the hour marked by an arrow. Variables were computed in hourly bins.
[0430] The following graphics convention have been used: All data are plotted as group mean ± SEM. The thick gray line encompasses the vehicle treatment mean ± SEM. Along the x-axis, time of treatment is marked by a triangle unless noted otherwise. Along the x-axis, light/dark bars indicate lights on/off.
Example 2. Effects of gabapentin on arousals, sleep continuity, and soporific efficacy
Gabapentin reduces number of arousals
[0431] The a2d-1 /a2d-2 ligand gabapentin administered orally to Wistar rats (N:=:9) at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), decreased the number of arousals, as demonstrated by an approximately 50% reduction in the number of wake bouts per hour post-treatment relative to vehicle control (as indicated by the arrow). Number of wake bouts plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment.
Statistically significant differences from methyicellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 1).
Gabapentin produces robust increases in sleep continuity/consolidation
[0432] The a2d-1/ a2d-2 ligand gabapentin administered orally to Wistar rats (N=9) at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation (as indicated by the arrow), as measured by the average sleep bout duration per hour. Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in a given hour for an individual animal, plotted as the population (gabapentin N 9; vehicle N=21) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant post-treatment differences from methylcellulose vehicle control within the initial 6 hours post-treatment are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa. At 100 mg/kg, gabapentin increased the average sleep bout >2-fold greater than vehicle controls (FIG. 2).
Gabapentin soporific efficacy as measured by EEG slow wave activity
[0433] The a2d-1/ a2d-2 ligand gabapentin administered to Wistar rats at CT-5 (5 hours after lights-on: time of treatment indicated by the triangle on the abscissa) increased EEG slow wave activity in nonREM sleep (as indicated by the arrow), as measured by the normalized (percent change from baseline) EEG delta power during nonREM sleep per hour. Normalized EEG delta power (power in the EEG at frequencies of 0.5-4.0 Hz, computed using Fourier analysis) is ploted in this example as the population (N=9) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle controls (N=2I) are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa. An increase in EEG slow wave activity is indicative of increased arousal threshold and depth of sleep due to the soporific effects of gabapentin (FIG. 3).
Example 3. Effects of pregabalin on number of arousals and sleep continuity
Pregahalin reduces number of arousals
[0434] The a2d-1 / a2d~2 ligand pregabalin (HY10204) administered to Wistar rats at CT~1 8 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), significantly decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts per hour, as compared to vehicle control treatment. Number of wake bouts are plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 4).
Pregabalin increases sleep continuity/consolidation
[0435] The a2d-!/ a2d-2 ligand pregabalin (HY- 10204) administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation as demonstrated by an >2-fold increase in average sleep bout duration per hour post-treatment (as indicated by the arrow) relative to vehicle control treatment. The robust sleep consolidating effect of pregabalin was approximately 2 times greater than the circadian peak for this measure during the undisturbed baseline circadian rest phase (circadian time 0:00-12:00). Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated m each hour for an individual animal, plotted as the population hourly mean ± SEM 30 hours before (baseline) and after treatment. Group sizes (N) are shown in the graph legend. Statistically significant differences from methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 5).
Example 4. Effects of trazadone on number of arousals and sleep continuity
Trazodone reduces number of arousals
[0436] The SHTIA antagonist trazodone administered to Wistar rats at CT-18 (6 hours after light- off; time of treatment indicated by the triangle on the abscissa), decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts, as compared to vehicle control treatment. Number of wake bouts are plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 6).
Trazodone increases sleep continuity/ consolidation
[0437] The 5HT2A antagonist trazodone administered to Wistar rats at CT-18 (6 hours after light- off; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation as demonstrated by the >2~fold increase in average sleep bout duration per hour post- treatment (as indicated by the arrow) relative to vehicle control treatment. Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in a given hour for an individual animal, plotted as the population (N=T2) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 7).
Example 5. Effects of nelotanseriii on number of arousals and sleep continuity
Nelotanserin reduces number of arousals [0438] The 5HT2A antagonist/inverse agonist nelotansenn administered to Wistar rats at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts, as compared to vehicle control treatment. Number of wake bouts are plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylceilulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 8).
Nelotanserin increases sleep continuity/consolidation
[0439] The 5HT?.A antagonist/inverse agonist nelotanserin administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation as demonstrated by the >2-fold increase in average sleep bout duration per hour post-treatment (as indicated by the arrow) relative to vehicle control treatment. Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in each hour for an individual animal, ploted as the population (N=8) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylceilulose vehicle control indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 9).
Example 6, Effects of pimavanserin on number of arousals and sleep continuity
Pimavanserin reduces number of arousals
[0440] The 5HT2A antagonist/inverse agonist pimavanserin administered to Wistar rats at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), significantly decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts per hour, as compared to vehicle control treatment. Effects were evident up to 7 hours post-treatment (arrow's). Number of wake bouts are plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from
methylceilulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 10).
Pimavanserin increases sleep continuity/consolidation
[0441] The 5HT2A antagonist/inverse agonist pimavanserin administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation as demonstrated by the approximately 2-fold increase in average sleep bout duration per hour post-treatment (as indicated by the arrow) relative to vehicle control treatment. Effects on sleep bout were long lasting at these doses, and particularly for the 30 mg/kg and 60 mg/kg doses where effect were still observed 28-30 hours after treatment. Pimavanserin 10 mg/kg effects on average sleep bout duration returned to vehicle treatment levels 18 hours after treatment. No further increase in average sleep bout duration was achieved with dose escalation beyond lOmg/kg, with the highest dose tested at 60 mg/kg. Thus, 10 mg/kg was the efficacy ceiling for pimavanserin. Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in a given hour for an individual animal, plotted as the group hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 11).
Example 7. Effects of doxepm on number of aronsals and sleep continuity
Doxepin reduces number oj iron sals
[0442] The histamine Hi receptor antagonist doxepin administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), decreased the number of arousals (as indicated by the arrow), as measured by the number of wake bouts, as compared to vehicle control treatment. Number of wake bouts are plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 12).
Doxepin increases sleep continuity /consolidation
[0443] The histamine Hi receptor antagonist doxepin administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), increased sleep consolidation as demonstrated by the approximately 2-fold increase in average sleep bout duration per hour post-treatment (as indicated by the arrow) relative to vehicle control treatment. Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in a given hour for an individual animal, plotted as the group (N=17) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 13).
Example 8. Effects of suvorexant on number of arousals and sleep continuity
Suvor exant fails to reduce the number of arousals
[0444] The orexin OX 1/0X2 antagonist suvorexant (HY-16387) administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), failed to reduce the number of arousals, as measured by the number of wake bouts. Instead, suvorexant increased the number of wake bouts. Number of wake bouts plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from vehicle control (TPGS) are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 14).
Weak effect of Suvorexant on sleep continuity/consolidation
[0445] The orexin OX 1/OX2 antagonist standard of care sedative hypnotic suvorexant (HY- 16387) administered to Wistar rats at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), produced a very weak increase in sleep consolidation (statistical trend 0.05<P< 1), as measured by the average sleep bout duration per hour (weak effect observed in the first two hours post-treatment; as indicated by the arrow). Average sleep bout duration was calculated as the mean duration of ail sleep bouts initiated in a given hour for an individual animal, plotted as the population (N:=:l 2) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 15).
Example 9. Effects of zolpidem on soporific efficacy, number of arousals, sleep continuity, REM sleep, and locomotor activity
Zolpidem soporific efficacy as measured by EEG slow wave activity
[0446] The standard of care treatment for insomnia, zolpidem, administered to Wistar rats at CT- 18 (6 hours after hghts-off; time of treatment indicated by the triangle on the abscissa) increased EEG slow wave activity in nonREM sleep (as indicated by the arrow), as measured by the normalized (percent change from baseline) EEG delta power during nonREM sleep per hour. Normalized EEG delta power (power in the EEG at frequencies of 0.5-4.0 Hz, computed using Fourier analysis) is ploted m this example as the population (N=17) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from
methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 16).
Soporific doses of zolpidem fail to reduce the number of arousals
[0447] The insomnia standard of care, zolpidem, administered at a dose that has robust soporific efficacy as measured by EEG slow wave activity' (FIG. 16) failed to reduce the number of arousals as measured by the number of wake bouts per hour. Zolpidem is a GABAA positive allosteric modulator acting at the benzodiazepine binding site on GABAA. In this example, Zolpidem (HY-10131) wras administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa). Number of wake bouts plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from vehicle control (methylcellulose) are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 17).
Small effect of zolpidem on sleep continuity/consolidation
[0448] At a dose that was sufficient to produce robust soporific efficacy as measured by EEG slow' wave activity' (FIG. 16), the benzodiazepine receptor mediated GABAA positive allosteric modulator, zolpidem (HY-10131), administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), produced only a small increase in sleep consolidation (statistical trend 0 Q5<P< 1), as measured by the average sleep bout duration per hour (weak effect observed in the first two hours post-treatment; as indicated by the arrow'). Average sleep bout duration was calculated as the mean duration of all sleep bouts initiated in a given hour for an individual animal, plotted as the population (N=8) hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from
methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 18).
Zolpidem inhibits REM sleep
[0449] The GABAA positive allosteric modulator, zolpidem (HY-10131) administered to Wistar rats at CT-5 (5 hours after light-on; time of treatment indicated by the triangle on the abscissa), inhibited REM sleep (as indicated by the arrow), as measured by the percent REM sleep that occurred per hour. The magnitude of REM sleep inhibition increases with dose and is severe in this zolpidem 30 mg/kg example. Data are plotted as the population hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from
methylcellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 19).
Zolpidem interferes with locomotor activity
[0450] The GABAA positive allosteric modulator sedative hypnotic, zolpidem (HY-1Q131), administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), inhibited Locomotor Activity Intensity (LMAi), as measured by the counts of locomotor activity per minute of wakefulness (as indicated by the arrow), averaged hourly for each animal. LMAi reveals changes in motor activity (movements) that are disproportionate from that normally observed during wakefulness. Hypoactivity during wakefulness, as shown here, is indicative of motor coordination impairment due to the well documented myorelaxant properties of zolpidem and related sedative hypnotics. The magnitude of undesirable motor impairment (reduction in LMAi) increases with dose, and is evident 4-5 hours post-treatment in this zolpidem 30 mg/kg example. Data are plotted as the population hourly mean ± SEM 30 hours before (baseline) and after treatment. Statistically significant differences from methylcellulose vehicle control are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 20).
Example 10. Effects of S-zopiclone on number of arousals, sleep continuity, locomotor activity, muscle tone, and rebound sleep disturbance
S-zopiclone failed to reduce the number of arousals
[0451] The insomnia standard of care, s-zopiclone, administered to Wistar rats at CT-18 (6 hours after light-off) failed to reduce the number of arousals, as measured by the number of wake bouts per hour post-treatment. Number of wake bouts are plotted as hourly mean ± SEM 30 hours before (baseline) and after treatment (red triangle on the abscissa). Statistically significant differences from vehicle control (methylcellulose) are indicated by asterisk. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 21).
Small (insufficient) effect of S-zopiclone on sleep continuity/consolidation.
[0452] Three doses of S-zopiclone (5 mg/kg. 10 mg/kg and 30 mg/kg) administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa). produced a weak increase in sleep consolidation that was statistically significant m the first treatment hour for the highest dose (30 mg/kg), as measured by the average sleep bout duration per hour (as indicated by the arrow). Effect on sleep consolidation was not dose proportional. Data are plotted as the population mean ± SEM each hour for 30 hours before (baseline) and after treatment. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa.
S-zopiclone dose-dependently interferes with locomotor activity
[0453] The insomnia standard of care sedative hypnotic s-zopiclone, administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), inhibited Locomotor Activity Intensity (LMAi) as measured by the counts of locomotor activity per minute of wakefulness, averaged hourly for each animal. LMAi reveals whether changes in motor activity (movements) are disproportionate from that normally observed during wakefulness. A disproportionate reduction in locomotor activity during wakefulness is shown. The reduction in LMAi increases with dose and is evident 1-6 hours post-treatment in the S- zopiclone 10 mg/kg and 30 mg/kg example (as indicated by the arrow). Data are plotted as the population mean ± SEM on an hourly basis for 30 hours before (baseline) and after treatment. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 1 : 12) is indicated on the abscissa (FIG. 23).
S-zopiclone decreases muscle tone as measured by electromyogram (EMG)
[0454] The insomnia standard of care sedative hypnotic s-zopiclone, administered to Wistar rats at CT-18 (6 hours after light-off), decreased EMG activity (as indicated by the arrow) as measured by root-mean-square assessment of EMG power in 5 minute intervals (treatment occurred at time zero). Data are plotted as population mean ± SEM. Statistically significant differences from methyleeliulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 24).
S-zopiclone soporific effects are followed by rebound sleep disturbance
[0455] The insomnia standard of care sedative hypnotic s-zopiclone, administered to Wistar rats at CT-18 (6 hours after light-off; time of treatment indicated by the triangle on the abscissa), initially increased the amount of nonREM (NREM) sleep post-treatment (as indicated by the arrow), but these initial soporific effects w¾re followed by an interval of sleep interference characterized by a reduction in sleep time, and corresponded with other markers of sleep disturbance such as concomitant reductions in sleep bout durations during the next circadian sleep phase (circadian time 0:00-12:00; FIG. 22). Data are plotted as the population mean ± SEM on an hourly basis for 30 hours before (baseline) and after treatment. Statistically significant differences from methy!cellulose vehicle control are indicated by asterisks. 24 hour light-dark cycle (LD 12: 12) is indicated on the abscissa (FIG. 25).
EQUIVALENTS
[0428] The details of one or more embodiments of the disclosure are set forth in the
accompanying description above. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, the preferred methods and materials are now described. Other features, objects, and advantages of the disclosure will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary' skill in the art to which this disclosure belongs. All patents and publications cited in this specification are incorporated by reference.
[0429] The foregoing description has been presented only for the purposes of illustration and is not intended to limit the disclosure to the precise form disclosed, but by the claims appended hereto.

Claims

1. A method of alleviating a symptom of, treating, or preventing a sleep disorder by administering a compound of the present disclosure, or pharmaceutically acceptable salt thereof, to a subject in need thereof, wherein the compound is a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a2d-2 auxiliary subunit of v oltage-gated calcium channels, a 5HT2A receptor antagonist, a SHIAA receptor inverse agonist, a H receptor antagonist, a Hi receptor inverse agonist, a gamma-hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or a pharmaceutically acceptable salt thereof.
2. The method of claim 1, wherein the sleep disorder is increased sleep fragmentation.
3. The method of claim 1 or 2, wherein the compound is selected from gabapentin, pregabalin, mirogaba!in, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
4. The method of any one of claims 1-3, wherein the compound is administered with one or more compounds selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof .
5. The method of any one of claims 1-4, wherein the compound is gabapentin, or a pharmaceutically acceptable salt thereof.
6. The method of any one of claims 1-4, wherein the compound is pregabalin, or a pharmaceutically acceptable salt thereof.
7. The method of any one of claims 1-4, wherein the compound is mirogabalin, or a pharmaceutically acceptable salt thereof.
8. The method of any one of claims 1-4, wherein the compound is trazodone, or a pharmaceutically acceptable salt thereof.
9. The method of any one of claims 1-4, wherein the compound is nelotanserm, or a pharmaceutically acceptable salt thereof.
10. The method of any one of claims 1-4, wherein the compound is pimavanserin, or a pharmaceutically acceptable salt thereof.
11. The method of any one of claims 1-4, wiierein the compound is doxepin, or a
pharmaceutically acceptable salt thereof.
12. The method of any one of claims 1-4, wherein the compound is diphenhydramine, or a pharmaceutically acceptable salt thereof.
13. The method of any one of claims 1-4, wherein the compound is gamma-hydroxy butyrate, or a pharmaceutically acceptable salt thereof.
14. The method of any one of the preceding claims, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respirator}- disturbance index
(RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal,
narcolepsy, mental disorder, or non-restorative sleep.
15. The method of claim 14, wherein the sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea, hypopnea, or acute hemoglobin desaturation.
16. The method of claim 14, wherein the neurological disease is Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or Lewy body dementia.
17. The method of claim 14, wherein the neurological disease is a neurodegenerative disease.
18. The method of claim 17, wherein the neurodegenerative disease is synucleinopathy.
19. The method of claim 18, wherein the synucleinopathy is Alzheimer’s disease,
Parkinson’s disease, or dementia with Lewy bodies
20. The method of claim 17, wherein the neurodegenerative disease is Lewy body disease, amyotrophic lateral sclerosis, Parkinson’s disease, Alzheimer’s disease, or Huntington’s disease.
21. The method of claim 14, wherein the pam is acute nociceptive pain, chronic neuropathic pain, inflammatory pain, arthritic pam, cancer pam, or mixed nociceptive and neuropathic pain.
22. The method of claim 21, wherein the mixed nociceptive and neuropathic pain is low back pain.
23. The method of claim 14, wherein the circadian rhythm disorder is jet-lag, shift-work, delayed sleep phase disorder, or non- 24 hour rhythm disorder.
24. The method of claim 14, wherein the substance abuse is opioid abuse or alcoholism.
25. The method of claim 14, wherein the substance abuse withdrawal is opioid withdrawal or alcohol withdrawal.
26. The method of claim 14, wherein the mental disorder is depression, major depressive disorder, post-traumatic stress disorder, anxiety disorder, bipolar disorder, or schizophrenia.
27. A compound of the present disclosure or a pharmaceutically acceptable salt thereof, for use in alleviating a symptom of, treating, or preventing a sleep disorder, wherein the compound is a ligand of a2d~1 auxiliary subunit of voltage-gated calcium channels, a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT?.A receptor antagonist, a 5HT2A receptor inverse agonist, a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma- hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or a pharmaceutically acceptable salt thereof.
28. The compound for use of claim 27, wherein the sleep disorder is increased sleep fragmentation.
29. The compound for use of claim 27 or 28, wherein the compound is selected from gabapentin, pregabalin, mirogabahn, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
30. The compound for use of any one of claims 27-29, wherein one or more compounds is selected from gabapentin, pregabalin, mirogabahn, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
31. The compound for use of any one of claims 27-30, wherein the compound is gabapentin, or a pharmaceutically acceptable salt thereof.
32. The compound for use of any one of claims 27-30, wherein the compound is pregabalin, or a pharmaceutically acceptable salt thereof.
33. The compound for use of any one of claims 27-30, wherein the compound is mirogabahn, or a pharmaceutically acceptable salt thereof.
34. The compound for use of any one of claims 27-30, wherein the compound is trazodone, or a pharmaceutically acceptable salt thereof
35. The compound for use of any one of claims 27-30, wherein the compound is nelotanserin, or a pharmaceutically acceptable salt thereof.
36. The compound for use of any one of claims 27-30, wherein the compound is pimavanserin, or a pharmaceutically acceptable salt thereof.
37. The compound for use of any one of claims 27-30, wherein the compound is doxepm, or a pharmaceutically acceptable salt thereof.
38. The compound for use of any one of claims 27-30, wherein the compound is diphenhydramine, or a pharmaceutically acceptable salt thereof.
39. The compound for use of any one of claims 27-30, wherein the compound is gamma- hydroxyhutyrate, or a pharmaceutically acceptable salt thereof
40. The compound for use of any one of claims 27-39, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respiratory disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal,
narcolepsy, mental disorder, or non-restorative sleep.
41. The compound for use of claim 40, wherein the sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea, hypopnea, or acute hemoglobin desaturation.
42. The compound for use of claim 40, wherein the neurological disease is Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or Lewy body dementia.
43. The compound for use of claim 40, wherein the neurological disease is a
neurodegenerative disease.
44. The compound for use of claim 43, wherein the neurodegenerative disease is synucleinopathy.
45. The compound for use of claim 44, wherein the synucleinopathy is Alzheimer’s disease, Parkinson’s disease, or dementia with Lewy bodies.
46. The compound for use of claim 43, wherein the neurodegenerative disease is Lewy body disease, amyotrophic lateral sclerosis, Parkinson’s disease, Alzheimer’s disease, or Huntington’s disease.
47. The compound for use of claim 40, wherein the pain is acute nociceptive pam, chronic neuropathic pam, inflammatory pam, arthritic pain, cancer pain, or mixed nociceptive and neuropathic pain.
48. The compound for use of claim 47, wherein the mixed nociceptive and neuropathic pain is low back pam.
49. The compound for use of claim 40, wherein the circadian rhythm disorder is jet- lag, shift work, delayed sleep phase disorder, or non-24 hour rhythm disorder.
50. The compound for use of claim 40, wherein the substance abuse is opioid abuse or alcoholism.
51. The compound for use of claim 40, wherein the substance abuse withdrawal is opioid withdrawal or alcohol withdrawal.
52. The compound for use of claim 40, wherein the mental disorder is depression, major depressive disorder, post-traumatic stress disorder, anxiety disorder, bipolar disorder, or schizophrenia.
53. Use of a compound of the present disclosure or a pharmaceutically acceptable salt thereof, for alleviating a symptom of, treating, or preventing a sleep disorder, wherein the compound is a ligand of a2d-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT2A receptor antagonist, a 5HT2A receptor inverse agonist, a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma- hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or a pharmaceutically acceptable salt thereof.
54. The use of claim 53, wherein the sleep disorder is increased sleep fragmentation.
55. The use of claim 53 or 54, wherein the compound is selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
56. The use of any one of claims 53-55, wherein one or more compounds is selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
57. The use of any one of claims 53-56, wherein the compound is gabapentin, or a pharmaceutically acceptable salt thereof.
58. The use of any one of claims 53-56, wherein the compound is pregabalin, or a pharmaceutically acceptable salt thereof.
59. The use of any one of claims 53-56, wherein the compound is mirogabalin, or a pharmaceutically acceptable salt thereof.
60. The use of any one of claims 53-56, wherein the compound is trazodone, or a pharmaceutically acceptable salt thereof.
61. The use of any one of claims 53-56, wherein the compound is nelotanserin, or a pharmaceutically acceptable salt thereof.
62. The use of any one of claims 53-56, wherein the compound is pimavanserin, or a pharmaceutically acceptable salt thereof.
63. The use of any one of claims 53-56, wiierein the compound is doxepin, or a
pharmaceutically acceptable salt thereof.
64. The use of any one of claims 53-56, wherein the compound is diphenhydramine, or a pharmaceutically acceptable salt thereof.
65. The use of any one of claims 53-56, wherein the compound is gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
66. The use of any one of claims 53-65, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respiratory' disturbance index (RDI), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
67. The use of claim 66, wherein the sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea, hypopnea, or acute hemoglobin desaturation.
68. The use of claim 66, wherein the neurological disease is Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or Lewy body dementia.
69. The use of claim 66, wherein the neurological disease is a neurodegenerative disease.
70. The use of claim 69, wherein the neurodegenerative disease is synucleinopathy.
71. The use of claim 70, wherein the synucleinopathy is Alzheimer’s disease, Parkinson’s disease, or dementia with Lewy bodies.
72. The use of claim 69, wherein the neurodegenerative disease is Lewy body disease, amyotrophic lateral sclerosis, Parkinson’s disease, Alzheimer’s disease, or Huntington’s disease.
73. The use of claim 66, wherein the pain is acute nociceptive pain, chronic neuropathic pain, inflammatory pain, arthritic pain, cancer pain, or mixed nociceptive and neuropathic pain.
74. The use of claim 73, wherein the mixed nociceptive and neuropathic pain is low back pain.
75. The use of claim 66, wherein the circadian rhythm disorder is jet-lag, shift- work, delayed sleep phase disorder, or non-24 hour rhythm disorder.
76. The use of claim 66, wherein the substance abuse is opioid abuse or alcoholism.
77. The use of claim 66, wherein the substance abuse withdrawal is opioid withdrawal or al coh ol with drawal .
78. The use of claim 66, wherein the mental disorder is depression, major depressive disorder, post-traumatic stress disorder, anxiety disorder, bipolar disorder, or schizophrenia.
79. Use of a compound of the present disclosure or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament, for alleviating a symptom of, treating, or preventing a ligand of «25-1 auxiliary subunit of voltage-gated calcium channels, a ligand of a2d-2 auxiliary subunit of voltage-gated calcium channels, a 5HT?A receptor antagonist, a 5HT2A receptor inverse agonist, a Hi receptor antagonist, a Hi receptor inverse agonist, a gamma- hydroxy butyrate (GHB) receptor ligand, or a GABAB receptor agonist, or a pharmaceutically acceptable salt thereof.
80. The use of a compound in the manufacture of a medicament of claim 79, wherein the sleep disorder is sleep fragmentation.
81. The use of a compound in the manufacture of a medicament of claim 79 or 80, wherein the compound is selected from gabapentm, pregabalin, mirogabalin, trazodone, nelotanserm, pimavanserin, doxepin, diphenhydramine, and gamma -hydroxy butyrate, or a pharmaceutically acceptable salt thereof.
82. The use of a compound in the manufacture of a medicament of any one of claims 79-81, wherein one or more compounds is selected from gabapentin, pregabalin, mirogabalin, trazodone, nelotanserin, pimavanserin, doxepin, diphenhydramine, and gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
83. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is gabapentin, or a pharmaceutically acceptable salt thereof.
84. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is pregabalin, or a pharmaceutically acceptable salt thereof.
85. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is mirogabalin, or a pharmaceutically acceptable salt thereof.
86. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is trazodone, or a pharmaceutically acceptable salt thereof.
87. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is nelotanserin, or a pharmaceutically acceptable salt thereof.
88. The use of a compound in the manufacture of a medicament of any one of claims 79-82, where the compound is pmiavanserm, or a pharmaceutically acceptable salt thereof.
89. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is doxepin, or a pharmaceutically acceptable salt thereof.
90. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is diphenhydramine, or a pharmaceutically acceptable salt thereof.
91. The use of a compound in the manufacture of a medicament of any one of claims 79-82, wherein the compound is gamma-hydroxybutyrate, or a pharmaceutically acceptable salt thereof.
92. The use of a compound in the manufacture of a medicament of any one of claims 79-91, wherein the sleep disorder is caused by or co-morbid with sleep apnea, restless legs syndrome, a high respiratory disturbance index (RD1), neurological disease, circadian rhythm disorder, pain, periodic leg movement disorder (PLMD), REM behavior disorder, elderly fragmented sleep, age-related sleep fragmentation, post-menopausal sleep disorder, substance abuse, substance abuse withdrawal, narcolepsy, mental disorder, or non-restorative sleep.
93. The use of a compound in the manufacture of a medicament of claim 92, wherein the sleep apnea is obstructive sleep apnea due to a high respiratory disturbance index (RDI) associated with an elevated respiratory event related arousal (RERA) with or without a concomitant apnea, hypopnea, or acute hemoglobin desaturation.
94. The use of a compound in the manufacture of a medicament of claim 92, wherein the neurological disease is Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or Lewy body dementia.
95. The use of a compound in the manufacture of a medicament of claim 92, wherein the neurological disease is a neurodegenerative disease.
96. The use of a compound in the manufacture of a medicament of claim 95, wherein the neurodegenerative disease is synucleinopathy.
97. The use of a compound in the manufacture of a medicament of claim 96, wherein the synucleinopathy is Alzheimer’s disease, Parkinson’s disease, or dementia with Lewy bodies.
98. The use of a compound in the manufacture of a medicament of claim 95, wherein the neurodegenerative disease is Lewy body disease, amyotrophic lateral sclerosis, Parkinson’s disease, Alzheimer’s disease, or Huntington’s disease.
99. The use of a compound in the manufacture of a medicament of claim 92, wherein the pain is acute nociceptive pain, chrome neuropathic pain, inflammatory pain, arthritic pam, cancer pain, or mixed nociceptive and neuropathic pam.
100. The use of a compound in the manufacture of a medicament of claim 99, wherein the mixed nociceptive and neuropathic pain is low back pain.
101. The use of a compound in the manufacture of a medicament of claim 92, wherein the circadian rhythm disorder is jet-lag, shift- work, delayed sleep phase disorder, or non-24 hour rhythm disorder.
102. The use of a compound in the manufacture of a medicament of claim 92, wherein the substance abuse is opioid abuse, or alcoholism.
103. The use of a compound in the manufacture of a medicament of claim 92, wherein the substance abuse withdrawal is opioid withdrawal or alcohol withdrawal.
104. The use of a compound in the manufacture of a medicament of claim 92, wherein the mental disorder is depression, major depressive disorder, post-traumatic stress disorder, anxiety disorder, bipolar disorder, or schizophrenia.
PCT/US2020/021770 2019-03-08 2020-03-09 Methods for treating sleep fragmentation disorders WO2020185712A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962815610P 2019-03-08 2019-03-08
US62/815,610 2019-03-08

Publications (1)

Publication Number Publication Date
WO2020185712A1 true WO2020185712A1 (en) 2020-09-17

Family

ID=72426746

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/021770 WO2020185712A1 (en) 2019-03-08 2020-03-09 Methods for treating sleep fragmentation disorders

Country Status (1)

Country Link
WO (1) WO2020185712A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110178166A1 (en) * 2006-05-19 2011-07-21 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110178166A1 (en) * 2006-05-19 2011-07-21 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep

Similar Documents

Publication Publication Date Title
WO2020185710A1 (en) Method for treating sleep fragmentation disorders using neurosteroid positive allosteric modulators of the gabaa receptor
Johnson et al. Sleep in children with autism spectrum disorders
US11690832B2 (en) Treatment of autoimmune diseases with combinations of RXR agonists and thyroid hormones
KR102149321B1 (en) Treatment of neurological disorders using a combination of RXR agonist and thyroid hormone
Sun et al. Effects of suvorexant, an orexin receptor antagonist, on breathing during sleep in patients with chronic obstructive pulmonary disease
Dumortier et al. Obstructive sleep apnea syndrome in adults with down syndrome: Causes and consequences. Is it a" chicken and egg" question?
WO2020185712A1 (en) Methods for treating sleep fragmentation disorders
AU2018425507B2 (en) Method of treatment with histamine-3 receptor inverse agonist
WO2020185711A1 (en) Method for treating sleep fragmentation disorders using dual acting h1 and 5ht2a inverse agonist/antagonists
Raymond et al. Quiet wakefulness: the influence of intraperitoneal and intranasal oxytocin on sleep–wake behavior and neurophysiology in rats
JP5779505B2 (en) New use of fibrates
Bae et al. Too Much of a Good Thing
NZ770420B2 (en) Method of treatment with histamine-3 receptor inverse agonist
Kennedy et al. A review of melatonin as a chronobiotic agent: therapeutic use in the treatment of sleep disorders in shift-workers

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: 20769014

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: 20769014

Country of ref document: EP

Kind code of ref document: A1