US20230093109A1 - Use of sublingual dexmedetomidine for the treatment of agitation - Google Patents

Use of sublingual dexmedetomidine for the treatment of agitation Download PDF

Info

Publication number
US20230093109A1
US20230093109A1 US17/990,332 US202217990332A US2023093109A1 US 20230093109 A1 US20230093109 A1 US 20230093109A1 US 202217990332 A US202217990332 A US 202217990332A US 2023093109 A1 US2023093109 A1 US 2023093109A1
Authority
US
United States
Prior art keywords
micrograms
agitation
dexmedetomidine
pharmaceutically acceptable
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/990,332
Inventor
Krishnan Nandabalan
Frank Yocca
Sameer Sharma
Harsh NEGI
Deepa Saini
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bioxcel Therapeutics Inc
Bioxcel LLC
Original Assignee
Bioxcel Therapeutics 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 Bioxcel Therapeutics Inc filed Critical Bioxcel Therapeutics Inc
Priority to US17/990,332 priority Critical patent/US20230093109A1/en
Assigned to E. Z. BIOXCEL SOLUTIONS PVT. LTD. reassignment E. Z. BIOXCEL SOLUTIONS PVT. LTD. INVENTION ASSIGNMENT AGREEMENT (EMPLOYMENT) Assignors: SAINI, DEEPA
Assigned to E. Z. BIOXCEL SOLUTIONS PVT. LTD. reassignment E. Z. BIOXCEL SOLUTIONS PVT. LTD. INVENTION ASSIGNMENT AGREEMENT (EMPLOYMENT) Assignors: NEGI, Harsh
Assigned to BIOXCEL THERAPEUTICS, INC. reassignment BIOXCEL THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: E. Z. BIOXCEL SOLUTIONS PVT. LTD.
Assigned to E. Z. BIOXCEL SOLUTIONS PVT. LTD. reassignment E. Z. BIOXCEL SOLUTIONS PVT. LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SHARMA, SAMEER
Assigned to BIOXCEL THERAPEUTICS, INC. reassignment BIOXCEL THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: YOCCA, FRANK
Assigned to BIOXCEL THERAPEUTICS, INC. reassignment BIOXCEL THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BIOXCEL CORPORATION
Assigned to BIOXCEL CORPORATION reassignment BIOXCEL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NANDABALAN, KRISHNAN
Publication of US20230093109A1 publication Critical patent/US20230093109A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/4174Arylalkylimidazoles, e.g. oxymetazolin, naphazoline, miconazole
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics

Definitions

  • the present invention discloses a method of treating agitation or the signs of agitation in a subject comprising sublingually administering an effective amount of an alpha-2 adrenergic agonist, more particularly Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • the present invention also discloses a sublingual composition for treating agitation or the signs of agitation comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients, along with the preparation thereof.
  • Agitation is an umbrella term that can refer to a range of behavioral disturbances or disorders, including aggression, combativeness, hyperactivity, and disinhibition. Agitation is a nonspecific constellation of relatively unrelated behaviors that can be seen in several different clinical conditions, usually presenting a fluctuating course. Agitation may be caused by several different medical conditions and drug interactions or by any circumstances that worsen the person's ability to think. Multiple underlying pathophysiologic abnormalities are mediated by dysregulations of dopaminergic, serotonergic, noradrenergic, and GABAergic systems. Agitation is characterized by non-productive, diffuse and excessive over-activity both motor (akathisia) and cognitive, and accompanied by an inner unpleasant tension. The key to safety is to intervene early to prevent progression of agitation to aggression and violence.
  • AD Alzheimer's disease dementia
  • FDD Fronto-temporal dementia
  • LBD Vascular dementia
  • LBD Lewy body disease
  • Down dementia Dementia in adults, gradually destroy a person's memory and ability to learn, reason, make judgments, communicate and carry out daily activities. In later stages, patients may experience changes in personality and behavior, such as anxiety, suspicion, agitation and aggression.
  • Agitation can also be associated with neuropsychiatric conditions such as schizophrenia, bipolar illness such as bipolar disorder or mania, depression, delirium, etc or agitation can be associated with alcohol and substance abuse withdrawal.
  • Acute agitation represented by a state of motor restlessness and accompanying mental tension, is a serious medical problem that can be present in some psychiatric disorders, including schizophrenia and bipolar mania, and may escalate quickly to aggressive behavior.
  • Acute agitation is characterized by signs that include pacing, hand wringing, fist clenching, pressured speech, yelling, and threatening people with escalated agitation.
  • beta blockers such as propranolol and Pindolol
  • anxiety medications such as Buspirone
  • benzodiazepines such as Lorazepam
  • anti-convulsants such as Valproate and Lamotrigine
  • anti-psychotics such as Haloperidol, Droperidol, Ziprasidone and other high-potency dopamine-blocking agents
  • atypical antipsychotics such as Olanzapine.
  • Buspirone, Valproate, Haloperidol, Droperidol and Ziprasidone have potential adverse effects, and optimal dosage and long-term efficacy in the management of chronic agitation in dementia is very limited.
  • Lorazepam is only effective for treating agitation in patients when used before medical procedures.
  • Loxapine an antipsychotic
  • Loxapine is FDA approved for treating agitated patients via inhalation, but is associated with a black box warning for bronchospasm and increased mortality in elderly patients with dementia-related psychosis (FDA label, Loxapine or Adasuve®).
  • Olanzapine, Ziprasidone or its combination with Haloperidol is also associated with QT prolongation, and extrapyramidal side effects should be watched very carefully in hospital set ups. Reports of adverse events (including eight fatalities) associated with intramuscular olanzapine underscores the need to follow strict prescribing guidelines and avoid simultaneous use with other CNS depressants.
  • an anti-agitation drug for schizophrenics or dementia patient should have a rapid onset of calming without sedation, be well tolerated and easy to administer with a high safety margin.
  • Alpha-2 adrenergic agonists have been used therapeutically for a number of conditions, including hypertension, congestive heart failure, angina pectoris, spasticity, glaucoma, diarrhea and for suppression of opiate withdrawal symptoms.
  • alpha-2 adrenergic agonists include Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine
  • (S)-4-[1-(2,3-Dimethylphenyl)ethyl]-3H-imidazole (Dexmedetomidine) is commercially available as an injectable formulation for sedation of initially intubated and mechanically ventilated patients during treatment in an intensive care setting, and for non-intubated patients prior to and/or during surgical and other procedures.
  • Dexmedetomidine is reported to have anti-agitational effects when administered intravenously or buccally during surgical procedures and intensive care unit (ICU) setups.
  • ICU intensive care unit
  • ICU intensive care unit
  • Ibacache et. al. in Anesthesia & Analgesia 2004 January; 98(1):60-3, discloses the administration of an intravenous single-dose of Dexmedetomidine to reduce agitation following sevoflurane anesthesia in children.
  • Other intravenous administrations are reported by Jeanne Boyer et al., in Nursing Critical care 2010 January, 5(1):30-34, Yahya Shehabi et. al., in Anesthetic Intensive Care 2010 January, 38(1):82-90, and Joseph D.
  • NCT 02720705 (clinical trial identification number from clinicaltrials.gov) discloses the administration of transbuccal Dexmedetomidine for the prevention of emergence agitation in pre-school children treated with sevoflurane in an intensive care unit setting.
  • Dexmedetomidine The sublingual use of Dexmedetomidine is disclosed in WO 2016/061413.
  • the focus of WO 2016/061413 is the administration of Dexmedetomidine sublingually at doses appropriate to treat sleep disorders and induce significant sedation.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof, administered sublingually can effectively treat agitation, including agitation associated with neurodegenerative diseases (e.g. Alzheimer's disease, fronto-temporal dementia, and sundown syndrome in Alzheimer's disease/dementia), agitation associated with neuropsychiatric conditions (e.g.
  • neurodegenerative diseases e.g. Alzheimer's disease, fronto-temporal dementia, and sundown syndrome in Alzheimer's disease/dementia
  • agitation associated with neuropsychiatric conditions e.g.
  • bipolar disorder schizophrenia, bipolar mania, delirium and depression
  • agitation associated with alcohol and substance abuse withdrawal or agitation associated with other conditions such as OPD/IPD procedures (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures).
  • OPD/IPD procedures e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures.
  • the dose to be administered sublingually may be selected to be effective to treat agitation, yet insufficient to causing significant sedation.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof sublingually to the subject, wherein the said agitation is associated with a neurodegenerative disease like dementia, Alzheimer's disease, frontotemporal dementia, or Parkinsonism, or associated with a neuropsychiatric condition like schizophrenia, bipolar disorder, bipolar mania, delirium, or depression, or associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), or associated with an alcohol and substance abuse withdrawal.
  • the agitation is suppressed without also causing significant sedation.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • Another aspect of the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with neurodegenerative disease, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • Yet another object of the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with dementia, Alzheimer's disease, frontotemporal dementia, Parkinsonism or other neurodegenerative diseases, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • Another object of the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, depression, or another related neuropsychiatric condition, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • a further object of the present invention provides a method of treating, preventing or reducing the signs of agitation in a subject in need thereof, wherein said agitation is associated with sundown syndrome in Alzheimer's disease/dementia, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • Yet another objective of the present invention provides a method for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an OPD/IPD procedure (e.g., MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • Yet another objective of the present invention provides a method for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an alcohol and substance abuse withdrawal, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • a further aspect of the present invention provides a sublingual composition for treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with a neurodegenerative disease, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutical acceptable carriers and/or excipients.
  • Another aspect of the present invention provides a sublingual composition for treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, depression, or another related neuropsychiatric condition, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically acceptable carriers and/or excipients.
  • An additional aspect of the present invention provides a sublingual composition for treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with sundown syndrome in Alzheimer's disease/dementia, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients.
  • Yet another aspect of the present invention provides a sublingual composition for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients.
  • OPD/IPD procedure e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures
  • Yet another aspect of the present invention provides a sublingual composition for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an alcohol and substance abuse withdrawal, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients.
  • Another object of the present invention provides a sublingual composition
  • a sublingual composition comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients, wherein said sublingual composition is selected from the group consisting of a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • a further object of the present invention provides a method of sublingually administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof to a subject's oral mucosa to treat agitation or the signs of agitation at a dosage which does not cause significant sedation.
  • the dosage administered sublingually may conveniently be in the range of between about 3 micrograms to about 100 micrograms
  • suitable dosages include: about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms.
  • the dose may be administered one or more times a day.
  • FIG. 1 A Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on cumulative duration of aggressive and agitated behaviors. Data expressed as Mean f SEM. One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 1 B Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on frequency of aggressive and agitated behaviors. Data expressed as Mean ⁇ SEM. One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 1 C Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on cumulative duration of aggressive and agitated behaviors. Data expressed as Mean ⁇ SEM. One-way ANOVA followed by Dunnett's post-hoc test. **p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 1 D Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on frequency of aggressive and agitated behaviors. Data expressed as Mean ⁇ SEM. One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 2 A Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Latency to attack. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 2 B Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Latency to attack. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 3 A Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Cumulative duration of Neutral behaviors such as grooming, and exploration. Data expressed as Mean f SEM. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 3 B Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Frequency of Neutral behaviors such as grooming, and exploration. Data expressed as Mean f SEM. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 3 C Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Neutral behaviors such as immobile/quiet time. Data expressed as Mean f SEM. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 3 D Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Cumulative duration of Neutral behaviors such as grooming, and exploration. Data expressed as Mean t SEM. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 3 E Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Frequency of Neutral behaviors such as grooming, and exploration. Data expressed as Mean ⁇ SEM. Data is expressed as Mean t SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 3 F Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 ⁇ g/kg) on Neutral behaviors such as immobile/quiet time. Data expressed as Mean ⁇ SEM. Data is expressed as Mean ⁇ SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p ⁇ 0.05 **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001 vs vehicle controls (vehicle).
  • Dex Dexmedetomidine hydrochloride
  • FIG. 4 A Mean plasma concentrations following Sublingual (SL) Dexmedetomidine hydrochloride administration in rats. Data expressed as Mean ⁇ SD
  • FIG. 4 B Mean plasma concentrations following Intravenous (IV) Dexmedetomidine hydrochloride administration in rats. Data expressed as Mean ⁇ SD
  • AD Alzheimer's disease
  • AUC Area under the curve
  • CNS Central nervous system CT/CAT scan: computed tomography scan
  • C max Maximum (or peak) serum concentration that a drug achieves in a specified compartment
  • EPS Extrapyramidal side effects
  • FTD Fronto-temporal dementia
  • GABA Gamma-aminoautyric Acid
  • ICU Intensive care unit
  • IPD In-Patient department
  • MRI Magnetic resonance imaging
  • OPD Out-patient department PTSD: Post-traumatic stress disorders
  • RSS Ramsay sedation score
  • RIT Rat intruder test
  • SLOS Smith-Lemli Opitz syndrome
  • T max Time at which the C max is observed.
  • treating refers to curative therapy, prophylactic therapy, and/or preventative therapy and can be used interchangeably.
  • composition As used herein, unless indicated otherwise, the terms “pharmaceutical composition”, “composition”, “formulation” and “composition of the invention,” are used interchangeably. Unless stated otherwise, the terms are meant to encompass, and are not limited to, pharmaceutical compositions containing drug substance i.e. Dexmedetomidine.
  • the composition may also contain one or more “excipients” that are “inactive ingredients” or “compounds” devoid of pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease or to affect the structure or any function of the human body.
  • an effective amount is interchangeable with “therapeutically effective dose,” or “therapeutically effective amount,” and refers to an amount sufficient to produce the desired effect. An effective amount is sufficient to cause an improvement in a clinically significant condition of the subject.
  • salt refers to a salt known to be non-toxic and commonly used in the pharmaceutical literature.
  • Typical inorganic acids used to form such salt include hydrochloric, hydrobromic, hydroiodic, nitric, sulfuric, phosphoric, hypophosphoric, and the like.
  • Salts derived from organic acids, such as aliphatic mono and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic and hydroxyl alkandioic acids, aromatic acids, aliphatic and aromatic sulfonic acids may also be used.
  • a preferred salt is the hydrochloride salt.
  • the term “subject” preferably refers to a human patient.
  • the subject can be any animal, including non-human mammals, such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates.
  • agitation means irritability, emotional outburst, impaired thinking, or excess motor and verbal activity that may occur due to either dysfunction of specific brain regions such as frontal lobes or due to dysfunction of neurotransmitter systems such as dopamine and nor-epinephrine.
  • agitation also includes aggression and hyper-arousal in post-traumatic stress disorder.
  • the agitation may be acute or chronic.
  • the signs of agitation includes excessive motor activity (examples include: pacing, rocking, gesturing, pointing fingers, restlessness, performing repetitious mannerisms), verbal aggression (e.g. yelling, speaking in an excessively loud voice, using profanity, screaming, shouting, threatening other people), physical aggression (e.g. grabbing, shoving, pushing, clenching hands into fists, resisting, hitting others, kicking objects or people, scratching, biting, throwing objects, hitting self, slamming doors, tearing things, and destroying property).
  • verbal aggression e.g. yelling, speaking in an excessively loud voice, using profanity, screaming, shouting, threatening other people
  • physical aggression e.g. grabbing, shoving, pushing, clenching hands into fists, resisting, hitting others, kicking objects or people, scratching, biting, throwing objects, hitting self, slamming doors, tearing things, and destroying property).
  • Acute agitation means agitation that occurs rapidly and is severe and sudden in onset. Acute agitation may be associated with, for example, neurodegenerative disease and neuropsychiatric conditions, although it may particularly exist in neuropsychiatric conditions. Acute agitation may lead to chronic agitation if it remains untreated.
  • chronic agitation means agitation developed over a long period of time, and is less severe than acute agitation. Chronic agitation may be associated with, for example, neurodegenerative disease and neuropsychiatric conditions, although it may particularly exist in neurodegenerative diseases.
  • neurodegenerative disease includes, but is not limited to, Alzheimer disease, frontotemporal dementia (or Pick's disease), Dementia, Dementia with Lewy bodies, post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, progressive supranuclear palsy or other related neurodegenerative diseases.
  • neuropsychiatric conditions includes, but is not limited to, schizophrenia, bipolar illness (bipolar disorder, bipolar mania), depression, delirium or other related neuropsychiatric conditions.
  • “Sundown syndrome” is a late-day circadian syndrome of increased confusion and restlessness, generally in a patient with some form of dementia. It seems to occur more frequently during the middle stages of Alzheimer dementia. It seems to subside with the progression of a patient's dementia. About 20-45% of Alzheimer type patients will experience some sort of sundowning confusion. Confusion and agitation worsen in the late afternoon and evening, or as the sun goes down.
  • perioperative agitation means agitation before, during or after any surgical procedure or ICU agitation unassociated with a neurodegenerative disease or neuropsychiatric condition.
  • sublingual literally means “under the tongue” and refers to a method of administering substances via the mouth in such a way that the substances are rapidly absorbed via the blood vessels under the tongue rather than via the digestive tract.
  • Sublingual absorption occurs through the highly vascularized sublingual mucosa, which allows a substance direct access to the blood circulation, thereby providing for direct systemic administration independent of gastrointestinal influences and avoiding undesirable first-pass hepatic metabolism. Accordingly, the total amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof in the formulation may be reduced, thereby reducing the likelihood of deleterious side effects and providing a cost benefit to the manufacturer.
  • “Sedation” as used herein means depressed consciousness in which a patient or subject retains the ability to independently and continuously maintain an open airway and a regular breathing pattern, and to respond appropriately and rationally to physical stimulation and verbal commands.
  • “without causing significant sedation” means that the patient experiences a level of sedation not greater than Level 3 on the Ramsay Sedation Scale. Level 3 means sedated but responds to commands.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject comprising administering an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • the alpha-2 adrenergic agonist includes, but is not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(
  • the present invention provides a method of treating agitation or the signs of agitation in a subject comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject.
  • the agitation is suppressed without also causing significant sedation.
  • Agitation may be effectively treated using a relatively low dose of Dexmedetomidine or a pharmaceutically acceptable salt thereof via the sublingual route. Consequently, in addition to providing relief from agitation without causing significant sedation, the treatment is also effective with reduced or no side effects (for example, cardiac or respiratory side effects).
  • the present invention is directed to a method of treating agitation or the signs of agitation in a subject comprising administering Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject to provide fast-acting relief without a substantial portion of Dexmedetomidine or its pharmaceutically acceptable salt thereof passing into the liver of the patient.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof via a sublingual composition to the subject, wherein the sublingual composition is selected from a film, wafer, patch, lozenge, gel, spray, tablet, and liquid drops.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to the subject an effective amount of an alpha-2 adrenergic agonist together with one or more pharmaceutically acceptable carriers and/or excipients via a sublingual composition, wherein the sublingual composition is a sublingual film.
  • the agitation is associated with a neurodegenerative disease or neuropsychiatric condition.
  • the treatment is effective without causing significant sedation.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to the subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients via a sublingual composition, wherein the sublingual composition is a sublingual film.
  • the agitation is associated with a neurodegenerative disease or neuropsychiatric condition.
  • the treatment is effective without causing significant sedation.
  • the present invention provides a method of treating agitation or signs of agitation in a subject in need thereof, comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof at a dosage that does not cause a significant sedation.
  • Suitable alpha-2 adrenergic agonists include, but are not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually at a dosage that does not cause significant sedation.
  • the dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof used in the sublingual composition is from about 3 micrograms to about 100 micrograms.
  • suitable dosages include: about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g.
  • the dose may be administered one or more times a day.
  • the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually at a dosage of from about 0.05 micrograms/kg weight of subject to about 1.5 micrograms/kg weight of subject.
  • suitable dosages include: about 0.1 micrograms/kg to about 1 micrograms/kg, about 0.1 micrograms/kg to about 0.5 micrograms/kg, about 0.1 micrograms/kg to about 0.4 micrograms/kg, about 0.1 micrograms/kg to about 0.3 micrograms/kg, about 0.1 micrograms/kg to about 0.2 micrograms/kg, about 0.07 micrograms/kg, about 0.05 micrograms/kg, about 0.1 micrograms/kg, about 0.2 micrograms/kg, about 0.3 micrograms/kg, about 0.4 micrograms/kg, about 0.5 micrograms/kg, about 0.6 micrograms/kg, about 0.7 micrograms/kg, about 0.8 micrograms/kg, about 0.9 micrograms/kg, about 1.0 micrograms/kg, about 1.1 micrograms/kg, about 1.2 micrograms/kg, about 1.3 micrograms/kg, about 1.4 micrograms/kg, about 1.5 micrograms/kg.
  • the dose may be administered one
  • the present invention provides a method of treating agitation or signs of agitation associated with neurodegenerative disease in a subject in need thereof, comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof at a dosage that does not cause a significant sedation.
  • Suitable alpha-2 adrenergic agonists include, but are not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-
  • the present invention provides a method of treating agitation or the signs of agitation associated with neurodegenerative disease in a subject in need thereof, comprising sublingually administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof at a dosage that does not cause unwanted (e.g., significant) sedation.
  • the dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof used may conveniently be from about 3 micrograms to about 100 micrograms, e.g.
  • micrograms to about 100 micrograms about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms, about 5 micrograms, about 6 micrograms, about 7 micrograms, about 8 micrograms, about 9 micrograms, about 10
  • the present invention provides a method of treating agitation or signs of agitation associated with neuropsychiatric condition in a subject in need thereof, comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof at a dosage that does not cause a significant sedation.
  • Suitable alpha-2 adrenergic agonists include, but are not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3
  • the present invention provides a method of treating agitation or the signs of agitation associated with neuropsychiatric condition in a subject in need thereof, comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually at a dosage that does not cause significant sedation.
  • the dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof used in a sublingual composition may conveniently be from about 3 micrograms to about 100 micrograms, e.g.
  • micrograms to about 100 micrograms about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms, about 5 micrograms, about 6 micrograms, about 7 micrograms, about 8 micrograms, about 9 micrograms, about 10
  • the level of acceptable sedation when treating a subject according to a method of the present invention is preferably at or below Level 3 according to the Ramsay sedation scoring (RSS) system.
  • RSS Ramsay sedation scoring
  • a particular embodiment of the present invention provides a method of treating agitation or the signs of agitation in a human subject in need thereof, comprising administering Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to said subject at a dose in the range of about 3 micrograms to about 100 micrograms, thereby achieving an RSS at or below Level 3 (e.g. Level 2 or Level 3).
  • the present invention also provides sublingual pharmaceutical compositions comprising an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof, preferably Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • the sublingual pharmaceutical compositions of the present invention may also comprise a pharmaceutically acceptable carrier and/or excipient.
  • suitable pharmaceutically acceptable carriers include water, sodium chloride, binders, penetration enhancers, diluents, lubricants, flavouring agents, coloring agents and so on.
  • compositions of the present invention may be administered to a subject alone or in combination with one or more other suitable active ingredients.
  • the present invention provides a sublingual pharmaceutical composition comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof for the treatment of agitation in a subject, e.g., agitation associated with neurodegenerative disease, sundown syndrome in Alzheimer's disease or dementia.
  • the sublingual pharmaceutical composition effectively treats agitation in a subject without causing significant sedation.
  • the present invention provides a sublingual pharmaceutical composition comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof for the treatment of agitation in a subject associated with schizophrenia, bipolar disorder, bipolar mania, other bipolar illness, depression, delirium or another related neuropsychiatric condition.
  • the sublingual pharmaceutical composition effectively treats agitation in a subject without causing significant sedation.
  • the sublingual pharmaceutical composition of the present invention may be, for example, a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • the sublingual pharmaceutical composition is in the form of a tablet or packed powder.
  • the sublingual pharmaceutical composition is in the form of a patch or film (e.g. thin film).
  • the patch may have adhesive qualities to prevent movement or swallowing of the patch.
  • the patch may be ingestible in case of accidental swallowing or to allow for its easy disposal, or the patch may be removed from under the tongue after a prescribed time.
  • the sublingual pharmaceutical composition is in the form of a paste, gel or ointment.
  • the viscosity of the paste, gel or ointment can be adjusted to allow for retention under the tongue.
  • the sublingual pharmaceutical composition is in a liquid (e.g. as a solution, suspension or emulsion), and may be, for example, presented as a spray or as drops.
  • Solutions include the active ingredient together with a diluent such as water, normal saline, sodium chloride solution, or any other suitable solvent such as propylene glycol, glycerol, ethyl alcohol and so on.
  • the diluent for the solution may particularly be physiological saline solution or water.
  • the amount of solution administered may conveniently be about 0.01 ml to about 1 ml (e.g., about 0.025-0.5 ml).
  • compositions of the invention may conveniently be administered by spraying, dripping, painting or squirting the composition under the tongue.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof is sublingually administered in liquid form, e.g. in a flavored or unflavored physiological saline solution.
  • the liquid composition may conveniently be administered under the tongue as drops or as a spray.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may conveniently represent from about 0.001% to about 99.99% of the overall composition, e.g. about 0.01% to about 90%, more particularly about 0.01% to about 30%.
  • a first unit dose is applied and held in place under the tongue for a predetermined time, for example for at least about 30 seconds, or more particularly about 60 seconds or more.
  • a second unit dose may then be applied and held in place for a similar amount of time.
  • the sublingual composition of Dexmedetomidine or a pharmaceutically acceptable salt thereof is a hard tablet or a compressed powder tablet.
  • the tablet may conveniently be designed to dissolve under the tongue in about 30 to 120 seconds as disclosed in U.S. Pat. No. 6,221,392 to Khankari, et al., incorporated herein by reference.
  • the sublingual composition of Dexmedetomidine or a pharmaceutically acceptable salt thereof is a hard tablet having a low grit component for an organoleptically pleasant mouth feel.
  • the tablet (or particles thereof containing the active ingredient which can be compressed to form the tablet) may also comprise a protective outer coating, e.g. any polymer conventionally used in the formation of microparticles, matrix-type microparticles and microcapsules.
  • the sublingual composition of Dexmedetomidine or a pharmaceutically acceptable salt thereof is a hard, compressed, rapidly dissolvable tablet.
  • the tablet conveniently includes the active ingredient within a matrix.
  • the matrix may be composed of, for example, at least one filler and a lubricant. Fillers include, for example, lactose or mannitol, and suitable lubricants include magnesium stearate, silicon dioxide and talc.
  • the matrix may also include one or more of: a binder (e.g. povidone, a sugar or carboxymethylcellulose), a disintegrant (e.g. croscarmellose sodium, crospovidone or sodium starch glycolate), a sweeting agent (e.g. sucralose) and the like.
  • the tablet may conveniently have a friability of about 2% or less and a hardness of about 15 to about 50 Newtons.
  • Another aspect of the present invention provides a method of making a packaged, sublingual tablet.
  • the method includes the steps of: (a) forming a mixture comprising Dexmedetomidine or a pharmaceutically acceptable salt thereof and a matrix including at least a non-direct compression filler and a lubricant; (b) compressing the mixture to form a plurality of hard, compressed, rapidly disintegrable particles (e.g. beads) including the active ingredient distributed in the sublingually dissolvable matrix; and (c) storing the product in bulk prior to packaging.
  • the dosage forms are then packaged in a lumen of a package such that there are more than one per package. Direct compression is the preferred method of forming the dosage forms.
  • an openable and reclosable package containing a plurality of hard, compressed, rapidly dissolving tablets adapted for direct oral dosing as described above.
  • the present invention is a sublingual tablet comprising an effervescent agent.
  • the effervescent agent may conveniently be present in an amount up to about 95% by weight, based on the weight of the finished tablet, and more particularly in an amount of between about 30% and about 80% by weight.
  • Sufficient effervescent material is included in the tablet composition to generate more than about 5 cm 3 but less that about 30 cm 3 of gas upon exposure of the tablet to an aqueous environment.
  • Sublingual compositions comprising effervescent agents are disclosed in U.S. Pat. No. 6,200,604, which is incorporated herein by reference.
  • an effervescent agent releases carbon dioxide e.g., as a result of the reaction of a soluble acid source with an alkaline carbonate or bicarbonate.
  • the acid source may conveniently include food acids and acids such as citric acid, tartaric, amalic, fumeric, adipic and succinic acid.
  • Carbonate and bicarbonate sources include dry solid carbonate and bicarbonate salts such as sodium bicarbonate, sodium carbonate, potassium bicarbonate, potassium carbonate, magnesium carbonate and the like.
  • Spray compositions of the present invention for sublingual administration may include one or more pharmaceutically acceptable liquids (e.g., present in the amount of about 30% to about 99.99% by weight of the composition).
  • Such liquids may be solvents, co-solvents, or non-solvents for Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • pharmaceutically acceptable liquids include water, ethanol, dimethyl sulfoxide, propylene glycol, polyethylene glycol, propylene carbonate, pharmaceutically acceptable oils (e.g., soybean, sunflower, peanut, peppermint etc.) and the like.
  • the pharmaceutically acceptable liquid is selected either to dissolve the active pharmaceutical ingredient, to produce a stable, homogenous suspension or solution of it, or to form any combination of a suspension or solution.
  • sublingual, spray formulations of Dexmedetomidine or a pharmaceutically acceptable salt thereof may include one or more carriers and/or excipients.
  • carriers/excipients include viscosity-modulating materials (e.g. polymers, sugars, sugar alcohols, gums, clays, silicas, and the like).
  • PVP polyvinylpyrrolidone
  • the viscosity-modulating material may conveniently be present in the amount of from about 0.01% to about 65% by weight of the spray formulation.
  • Other examples of carriers/excipients include preservatives (e.g. ethanol, benzyl alcohol, propylparaben and methylparaben).
  • Preservatives may conveniently be present in the amount of from about 0.001% to about 10% by weight of the spray formulation.
  • Carriers/excipients may also be flavoring agents, sweeteners (e.g. sugars such as sucrose, glucose, dextrose, maltose, fructose, etc.), artificial sweeteners (e.g. saccharin, aspartame, acesulfame, sucralose etc.), or sugar alcohols (e.g. mannitol, xylitol, lactitol, maltitol syrup etc.) present conveniently in an amount of from about 0.001% to about 65% by weight of the spray formulation.
  • sweeteners e.g. sugars such as sucrose, glucose, dextrose, maltose, fructose, etc.
  • artificial sweeteners e.g. saccharin, aspartame, acesulfame, sucralose etc.
  • sugar alcohols e.g. mannitol,
  • carriers/excipients include buffers and pH-adjusting agent (e.g., sodium hydroxide, citrate, and citric acid) conveniently present in an amount of from about 0.01% to about 5% by weight of the spray formulation.
  • Coloring agents e.g. present in an amount of from about 0.001% to about 5% by weight of the spray formulation
  • fragrances e.g. present in an amount of from about 0.001% to about 1% by weight of the spray formulation
  • chelating agents such as EDTA (e.g. present in an amount of from about 0.001% to about 1% by weight of the spray formulation)
  • UV absorbers e.g. present in an amount of from about 0.001% to about 10% by weight of the spray formulation
  • anti-foam agents e.g. low molecular weight alcohols, dimethicone
  • One particular aspect of the present invention provides a sublingual film comprising Dexmedetomidine or a pharmaceutically acceptable salt thereof, together with one or more carriers and/or excipients, for the treatment of agitation.
  • Excipients which may be incorporated into the sublingual films of the present invention include one or more of the following: film forming agents, mouth feel improvers, plasticizers, stabilizers, surfactants, preservatives, sweetening agents, colorants, flavourants, emulsifiers, disintegrants, salivating agents, antioxidants, permeation enhancers, solvents and the like.
  • Film forming agents generally mean agents that provide structure to the film of the present invention.
  • the effective amount of the film forming agent ranges from about 10% to about 99%, more preferably about 50% to about 90% by weight of the composition.
  • Film forming agents that can be utilized as part of the film composition of the present invention include, but are not limited to, cellulose ethers, modified starches, natural gums, edible polymers, seaweed extracts, land plant extracts, pullulan, polyvinylpyrrolidone, derivatives thereof and combinations thereof.
  • cellulose ethers include, but are not limited to, methylhydroxycellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, carboxymethylcellulose, derivatives thereof and combinations thereof.
  • Modified starches include, but are not limited to, acid and enzyme hydrolyzed corn and potato starches, derivatives thereof and combinations thereof.
  • natural gums include, but are not limited to, gum arabic, guar gum, locust bean gum, carrageenan gum, acacia, karaya, ghatti, tragacanth agar, tamarind gum, xanthan gum, derivatives thereof and combinations thereof.
  • edible polymers include, but are not limited to, microcrystalline cellulose, cellulose ethers, xanthan, derivatives thereof and combinations thereof.
  • Seaweed extract examples include, but are not limited to, sodium alginate, carrageenans, derivatives thereof and combinations thereof.
  • Land plant extracts include, but are not limited to, konjac, pectin, arabinogalactan, derivatives thereof and combinations thereof.
  • Particular film forming agents include pullulan, sodium alginate, polyvinylpyrrolidone, methylcellulose and methylhydroxycellulose (MHC).
  • solvent generally refers to liquids that will dissolve solutes.
  • a solvent may be used to dissolve film-forming agents and other excipients to prepare film-forming compositions of the present invention.
  • Solvents include, but are not limited to, demineralized/distilled water, ethyl alcohol, isopropyl alcohol, methyl ethyl ketone, propylene glycol methyl ether acetate, dimethyl acetamide, ethylene glycol mono-propyl ether, and toluene.
  • a sublingual film of the present invention may conveniently comprise a solvent in an amount up to about 1% w/w.
  • stabilizer generally refers to an agent that will impart stability to the formulation during its shelf life.
  • Stabilizers of the present invention can include, for example, oil/water emulsifiers and flavor fixatives.
  • the effective amount of a stabilizer agent in a composition of the invention may be, for example, in the range of about 0% to about 45%, more particularly about 4% to about 25%, by weight of the composition.
  • suitable stabilizing agents of the present invention include, but are not limited to, gum arabic, microcrystalline cellulose, carrageenan, xanthan gum, locust bean gum, derivatives thereof and combinations thereof.
  • Particular stabilizing agents of the present invention include gum arabic and microcrystalline cellulose.
  • Disintegrants can aid the dissolution of edible films allowing for the efficacy of the film to be realized sooner.
  • Suitable disintegrants for use in an edible film of the present invention include, but are not limited to, alginic acid, microcrystalline cellulose and carboxymethylcellulose.
  • Special disintegrants known as super-disintegrants are also suitable for use in an edible film of the present invention.
  • Super-disintegrants include cross-linked polymers (e.g. crospovidone), cross-linked starches (e.g. sodium starch glycolate), and cross-linked celluloses (e.g. a modified carboxymethylcellulose such as croscarmellose).
  • the disintegrants or super-disintegrants are conveniently present in a sublingual composition of this invention (e.g. an edible film) in an amount ranging from about 1% to about 10%, more particularly about 1% to about 5% by weight of the composition.
  • a sublingual composition of this invention e.g. an edible film
  • Emulsifiers suitable for use in an edible film of the present invention include, but are not limited to, gum arabic, carrageenan, triethanolamine stearate, quaternary ammonium compounds, acacia, gelatin, lecithin, bentonite, veegum, derivatives thereof and combinations thereof.
  • Emulsifiers can be used in a composition of the present invention in an amount up to about 40%, more particularly up to about 25%, by weight of the composition.
  • the emulsifier can be a stabilizer creating an oil/water emulsion encapsulating volatile oils and flavoring agents, thereby essentially acting as a flavor fixative.
  • a particular emulsifier for use in an edible film of the present invention is gum arabic.
  • plasticizing agent or “plasticizer” may be utilized to improve flexibility and reduce brittleness of an edible film composition of the present invention.
  • the plasticizing agent may conveniently constitute up to about 30%, e.g. up to about 15% by weight of the composition.
  • suitable plasticizing agents include, but are not limited to, glycerin, sorbitol, triacetin, monoacetin, diacetin, polyethylene glycol, propylene glycol, hydrogenated starch hydrolysates, corn syrups, low molecular weight propylene glycols, phthalate derivatives like dimethyl, diethyl and dibutyl phthalate, citrate derivatives such as tributyl, triethyl, acetyl citrate and castor oil derivatives thereof and combinations thereof.
  • Particular plasticizing agents of the present invention include sorbitol and glycerin.
  • preservative generally refers to an excipient used to kill microorganisms or prevents, inhibits or retards their growth and reproduction, and is included in a product in a concentration only sufficient to prevent spoilage or the growth of inadvertently added microorganisms.
  • Suitable preservative includes, but are not limited to, methylparaben, propylparaben and sodium benzoate.
  • the preservative may conveniently be present in the composition from about 0.001% to about 10% w/w of the composition.
  • sweetening agent generally refers to an excipient used to impart sweetness to a pharmaceutical composition.
  • suitable sweetening agents for use in a composition of the present invention include, but are not limited to, aspartame, dextrose, glycerin, mannitol, saccharin sodium, sorbitol and sucrose.
  • the sweetening agent may conveniently be present in the composition in an amount of from about 5% to about 20% w/w of the composition.
  • coloring agent generally refers to an excipient used to impart color to a pharmaceutical composition. Suitable colorants include, but are not limited to, FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, other F.D. & C. dyes, caramel, red ferric oxide, and natural coloring agents such as grape skin extract, beet red powder, beta-carotene, annatto, carmine, turmeric or paprika.
  • the colorant may conveniently be present in the composition in an amount of from about 0.001% to about 10% w/w of the composition.
  • flavoring agent or “flavorant” generally refers to an excipient used to impart a pleasant flavor (and often also odor) to a pharmaceutical composition.
  • suitable flavorants include, but are not limited to, synthetic flavoring oils, flavoring aromatics, natural oils, extracts from whole plants or parts thereof such as leaves, flowers, fruits or combinations thereof. Examples include cinnamon oil, wintergreen oil, peppermint oil, clove oil, bay oil, anise oil, eucalyptus oil, thyme oil, cedar leave oil, nutmeg oil, sage oil, bitter almond oil and cassia oil.
  • flavorants include vanilla, citrus fruit oils such as lemon, orange, grape, lime or grapefruit oil, and fruit essences such as apple, pear, peach, strawberry, raspberry, cherry, plum, pineapple or apricot essence.
  • Flavorants of particular interest for use in a composition of the present invention include commercially available orange, grape, cherry and bubble gum flavors and mixtures thereof. The amount of flavoring used will depend on a number of factors, including the organoleptic effect desired. Particular flavorants include grape and cherry flavors, and citrus fruit flavors such as orange flavor. The flavorant may conveniently be present in the composition in an amount of from about 0.001% to about 10% w/w of the composition.
  • salivating agent is an agent that promotes greater salivation during use of a composition of the present invention. This may be an important feature if the composition is intended to be taken by the patient without the aid of water to help in the transporting of the composition to the stomach of the patient.
  • the salivating agent can be, for example, an emulsifier or a food acid that initiates salivation in the mouth of the patient.
  • emulsifiers useful as salivating agents include alkyl aryl sulfonates, alkyl sulfates, sulfonated amides and amines, sulfated and sulfonated esters and ethers, alkyl sulfonates, polyethoxylated esters, mono-, di-, and triglycerides, diacetyl tartaric esters of monoglycerides, polyglycerol esters, sorbitan esters and ethoxylates, lactylated esters, phospholipids such as lecithin, polyoxyethylene sorbitan esters, propylene glycol esters, sucrose esters, and mixtures thereof.
  • the emulsifier may be either saturated or unsaturated. It should be noted that some of the emulsifiers that are salivating agents may also function as binders. Examples of food acids useful as salivating agents include citric acid, malic acid, tartarate, food salts such as sodium chloride and salt substitutes, potassium chloride, and mixtures thereof.
  • the amount of salivating agent present in a sublingual film of the present invention may convenient be up to about 15% by weight of the final composition, e.g. in the range of from about 0.3% to 0.4% by weight of the composition.
  • antioxidant generally refers to an excipient used to inhibit oxidation and thus prevent deterioration of active agents by oxidative processes.
  • Suitable antioxidants include, for example, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothio-glycerol, propyl gallate, sodium ascorbate, citric acid, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, EDTA and sodium edetate.
  • the anti-oxidant may conveniently be present in the composition in an amount of from about 0.001% to about 2% w/w of the composition.
  • permeation enhancer generally refers to an excipient used to enhance permeation of an active agent to cellular membranes or enhance the local/systemic absorption of the active agent.
  • Permeation enhancers that may be used in the present invention include, but are not limited to, solubilizers such as alcohols, polyethylene glycols, chelating agents (e.g. cyclodextrins), sucrose laurate or sucrose oleate.
  • the permeation enhancer may conveniently be present in the composition in an amount of from about 0.1% to about 5% w/w of the composition.
  • the sublingual pharmaceutical composition of the present invention includes a mucosal permeation enhancer appropriate for enhancing the mucosal absorption of the composition.
  • Sublingual Dexmedetomidine formulations may be made by mixing appropriate quantities of the foregoing ingredients in accordance with standard good manufacturing practices. The relative amounts of each ingredient should not interfere with the desirable pharmacological and pharmacokinetic properties of the resulting formulation.
  • Sublingual Dexmedetomidine films of the present invention may be conveniently prepared using PharmFilm® technology (owned by MonoSol) or technology owned by ARx LLC.
  • PharmFilm® technology owned by MonoSol
  • ARx LLC Various patents and patent applications are incorporated herein in entirety and includes U.S. Pat. Nos. 9,585,961, 7,470,397, 7,727,466, 9,248,146, 9,545,376, 2017-0087084, 9,662,297, 9,662,301, 2017-0246108, 2017-0252294, 9,441,142 assigned to ARx LLC and U.S. Pat. Nos.
  • the active agent e.g. Dexmedetomidine or a pharmaceutically acceptable salt thereof
  • film forming agents and optionally one or more carriers and/or excipients selected from the group comprising of mouth feel improver, plasticizer, stabilizer, surfactant, preservative, sweetening agent, colorant, flavourant, emulsifier, disintegrant, salivating agent, antioxidant, permeation enhancer are dissolved in a compatible solvent to form a film forming composition.
  • Compatible solvents include water, alcohols such as ethanol, ethyl acetate, acetone, and mixtures thereof.
  • the film forming composition is cast on a releasable carrier and dried to form a sheet/film.
  • the carrier material must have a surface tension which allows the film solution to spread evenly across the intended carrier width without soaking to form a destructive bond between the film carrier substrates.
  • suitable carrier materials include glass, stainless steel, Teflon and polyethylene-impregnated paper. Drying of the film may be carried out at high temperature using a drying oven, drying terminal, vacuum drier, or any other suitable drying equipment which does not adversely affect the ingredients of which the film is composed.
  • the sublingual film of the present invention can also be prepared by other established processes e.g. extrusion (for example, Hot melt extrusion, Solid dispersion extrusion), casting (for example, solid casting or semi-solid casting), Rolling methods and the like.
  • the present invention provides a sublingual composition
  • a sublingual composition comprising an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof, administered to a subject in an amount sufficient to effectively treat agitation.
  • the amount of alpha-2 adrenergic agonist is sufficient to effectively treat agitation without causing significant sedation.
  • the alpha-2 adrenergic agonist may conveniently be delivered on an “as needed basis” in one, two or more doses per day to the animal (e.g. human) subject.
  • the composition may also be administered via a single dosage form or via multiple dosage forms.
  • the present invention provides a sublingual composition
  • a sublingual composition comprising Dexmedetomidine or a pharmaceutically acceptable salt thereof, administered to a subject in an amount sufficient to effectively treat agitation.
  • the amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof used is sufficient to effectively treat agitation without causing significant sedation.
  • the Dexmedetomidine or a pharmaceutically acceptable salt thereof may conveniently be delivered on an “as needed basis” in one, two or more doses per day to the animal (e.g. human) subject.
  • the composition may also be administered via a single dosage form or via multiple dosage forms.
  • a therapeutic (i.e. anti-agitation) effect may begin within about 60 minutes (e.g. within about 30, 20, 15, 10, 5, 3, 2 or 1 minutes) after administration, or within about 30 seconds after administration.
  • the signs of agitation may also be relieved within about 1 to about 60 minutes after administration, and more typically within about 5 to about 30 minutes.
  • a second dose of the composition of this invention may be administered to the subject if the signs of agitation are not relieved within about 60 minutes.
  • Treatment protocols may include one or more dosage intervals (e.g. two or more dosage intervals, five or more dosage intervals, or ten or more dosage intervals). Depending on the physiology of the subject and the desired therapeutic effect, the duration of dosage intervals and treatment protocols according to embodiments of the present invention may vary.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may be administered as a sublingual composition to treat agitation or the signs of agitation either alone or in combination with one or more further active agents.
  • the active agents can either be formulated as a single composition or as two or more separate compositions, which can be administered simultaneously, sequentially or separated by an appropriate period of time.
  • the weight ratio of respectively Dexmedetomidine or a pharmaceutically acceptable salt thereof to the second active agent may generally be in the range from about 1:2 to about 1:2.5; about 1:2.5 to about 1:3; about 1:3 to about 1:3.5 about 1:3.5 to about 1:4; about 1:4 to about 1:4.5, about 1:4.5 to about 1:5; about 1:5 to about 1:10; and about 1:10 to about 1:25.
  • the weight ratio may particularly be between about 1:1 to about 1:5; about 1:5 to about 1:10; about 1:10 to about 1:15; or about 1:15 to about 1:25.
  • the weight ratio of respectively the second active agent to Dexmedetomidine or a pharmaceutically acceptable salt may be in the range of from about 2:1 to about 2.5:1; about 2.5:1 to about 3:1; about 3:1 to about 3.5:1; about 3.5:1 to about 4:1; about 4:1 to about 4.5:1; about 4.5:1 to about 5:1; about 5:1 to about 10:1; and about 10:1 to about 25:1.
  • the weight ratio of respectively the second active agent to Dexmedetomidine or a pharmaceutically acceptable salt thereof may particularly be in the range of from about 1:1 to about 5:1; about 5:1 to about 10:1; about 10:1 to about 15:1; or about 15:1 to about 25:1. It is to be understood that all ranges between the quoted ranges are also covered herein, and constitute further particular aspects of this invention.
  • the dosing regimen employed may depend on several factors, such as the type of agitation treated, the severity of the signs, and whether the agitation is due to an underlying medical condition.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may be administered sublingually in any appropriate dose to an animal (e.g. human).
  • the human dose may be from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about
  • the dose may be administered one or more times a day.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may be administered sublingually in any appropriate dose to a human.
  • the human dose may be from about 0.05 micrograms/kg weight of subject to about 1.5 micrograms/kg weight of subject.
  • suitable dosages include: about 0.1 micrograms/kg to about 1 micrograms/kg, about 0.1 micrograms/kg to about 0.5 micrograms/kg, about 0.1 micrograms/kg to about 0.4 micrograms/kg, about 0.1 micrograms/kg to about 0.3 micrograms/kg, about 0.1 micrograms/kg to about 0.2 micrograms/kg, about 0.07 micrograms/kg, about 0.05 micrograms/kg, about 0.1 micrograms/kg, about 0.2 micrograms/kg, about 0.3 micrograms/kg, about 0.4 micrograms/kg, about 0.5 micrograms/kg, about 0.6 micrograms/kg, about 0.7 micrograms/kg, about 0.8 micrograms/kg, about 0.9 micrograms
  • Embodiment 1 A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering sublingually to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • Embodiment 2 A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, wherein said Dexmedetomidine or a pharmaceutically acceptable salt thereof is sublingually administered at a dosage that treats agitation or the signs of agitation without causing significant sedation.
  • Embodiment 3 The method according to Embodiment 1 or 2, wherein said dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof is in range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms
  • micrograms about 5, 6, 7, 8, or 9 micrograms
  • about 10 micrograms about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 4 The method according to Embodiments 1, 2 or 3, wherein said subject is mammal, preferably human.
  • Embodiment 5 The method according to any one of Embodiments 1 to 4, wherein said agitation is associated with a neurodegenerative disease.
  • Embodiment 6 The method according to Embodiment 5, wherein said neurodegenerative disease is selected from Alzheimer disease, fronto-temporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, and progressive supranuclear palsy.
  • FTD fronto-temporal dementia
  • DLB dementia with Lewy bodies
  • Parkinson's disease vascular dementia
  • vascular cognitive impairment Huntington's disease
  • multiple sclerosis Creutzfeldt-Jakob disease
  • progressive supranuclear palsy progressive supranuclear palsy.
  • Embodiment 7 The method according to any one of Embodiments 1 to 4, wherein the agitation is associated with a neuropsychiatric condition.
  • Embodiment 8 The method according to Embodiment 7, wherein said neuropsychiatric condition is selected from schizophrenia, bipolar illness (such as mania, disorder), delirium, and depression.
  • Embodiment 9 The method according to Embodiment 5, wherein the agitation is associated with sundown syndrome in dementia or Alzheimer's disease.
  • Embodiment 10 A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is not perioperative agitation and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 11 A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with a neurodegenerative disease and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 12 A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with a neuropsychiatric condition and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 13 A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with sundown syndrome in dementia or Alzheimer's disease and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 14 The sublingual composition according to Embodiment 11, wherein said neurodegenerative disease is selected from the group consisting of Alzheimer disease, frontotemporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis Creutzfeldt-Jakob disease, multiple system atrophy, and progressive supranuclear palsy.
  • FDD frontotemporal dementia
  • DLB dementia with Lewy bodies
  • Parkinson's disease vascular dementia
  • vascular cognitive impairment Huntington's disease
  • multiple sclerosis Creutzfeldt-Jakob disease multiple system atrophy
  • progressive supranuclear palsy progressive supranuclear palsy.
  • Embodiment 15 The sublingual composition according to Embodiment 14, wherein said neurodegenerative disease is selected from dementia, frontotemporal dementia, Alzheimer's disease and Parkinson's disease.
  • Embodiment 16 The sublingual composition according to Embodiment 12, wherein said neuropsychiatric condition is selected from the group consisting of schizophrenia, bipolar illness (such as mania, disorder), delirium and depression.
  • Embodiment 17 The sublingual composition according to any one of Embodiments 10 to 16, wherein said composition is selected from a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • Embodiment 18 The sublingual composition according to Embodiment 17, wherein the composition is a film.
  • Embodiment 19 The sublingual composition according to Embodiment 18, wherein the film is mucoadhesive in nature and provides a quick onset of action.
  • Embodiment 20 The sublingual composition according to any one of Embodiments 10 to 19, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered at a dosage that treats agitation or the signs of agitation without causing significant sedation.
  • Embodiment 21 The sublingual composition according to Embodiment 20 wherein the observed level of sedation is not greater than 3 on the Ramsay sedation scale.
  • Embodiment 22 The sublingual composition according to any one of Embodiments 10 to 21, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered to said subject (e.g. human) at a dosage in the range of from about 3 micrograms to about 100 micrograms (e.g.
  • micrograms to about 100 micrograms about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g.
  • micrograms about 5, 6, 7, 8, or 9 micrograms
  • about 10 micrograms about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 23 A method of treating agitation or the signs of agitation in a subject in need thereof, the method comprises sublingually administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, wherein the agitation is not perioperative agitation.
  • Embodiment 24 The method according to Embodiment 23, wherein the agitation is associated with a neurodegenerative disease and/or neuropsychiatric condition.
  • Embodiment 25 The method according to Embodiment 24, wherein the neurodegenerative disease is selected from the group consisting of Alzheimer disease, frontotemporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, progressive supranuclear palsy or other related neurodegenerative disorder.
  • FDD frontotemporal dementia
  • DLB dementia with Lewy bodies
  • post-traumatic stress disorder Parkinson's disease
  • vascular dementia vascular cognitive impairment
  • Huntington's disease multiple sclerosis
  • Creutzfeldt-Jakob disease multiple system atrophy
  • progressive supranuclear palsy or other related neurodegenerative disorder is selected from the group consisting of Alzheimer disease, frontotemporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment
  • Embodiment 26 The method according to Embodiment 24, wherein the neuropsychiatric condition is selected from the group consisting of schizophrenia, bipolar illness (e.g. bipolar disorder or bipolar mania), delirium and depression.
  • the neuropsychiatric condition is selected from the group consisting of schizophrenia, bipolar illness (e.g. bipolar disorder or bipolar mania), delirium and depression.
  • Embodiment 27 The method according to any one of Embodiments 23 to 26, wherein agitation or the signs of agitation are treated effectively without causing significant sedation.
  • Embodiment 28 The method according to any one of Embodiments 23 to 27, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered in form of a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • Embodiment 29 A method of treating of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), and said method comprises administering to said subject sublingually an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • OPD/IPD procedure e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures
  • Embodiment 30 A method of treating of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with an alcohol and substance abuse withdrawal and said method comprises administering to said subject sublingually an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • Embodiment 31 The method according to Embodiment 29 or 30, wherein said Dexmedetomidine or a pharmaceutically acceptable salt thereof is sublingually administered at a dosage that treats said agitation or the signs of agitation without causing significant sedation.
  • Embodiment 32 The method according to Embodiment 31, wherein said dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof is in range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about
  • micrograms about 5, 6, 7, 8, or 9 micrograms
  • about 10 micrograms about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 33 The composition or method according to any preceding Embodiment, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered once, twice or thrice daily or on an “as needed” basis.
  • Embodiment 34 The composition or method according to any preceding Embodiment, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered in a manner that produces a therapeutic effect in less than about 60 minutes, particularly within about 30 seconds to about 30 minutes.
  • Embodiment 35 A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering sublingually to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof.
  • Embodiment 36 A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof, wherein said alpha-2 adrenergic agonist is sublingually administered at a dosage that treats agitation or the signs of agitation without causing significant sedation.
  • Embodiment 37 The method according to Embodiment 35 or 36, wherein said dosage of alpha-2 adrenergic agonist is in range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15
  • micrograms about 5, 6, 7, 8, or 9 micrograms
  • about 10 micrograms about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 38 The method according to Embodiments 35, 36 or 37, wherein said subject is mammal, preferably human.
  • Embodiment 39 The method according to any one of Embodiments 35 to 38, wherein said agitation is associated with a neurodegenerative disease.
  • Embodiment 40 The method according to Embodiment 39, wherein said neurodegenerative disease is selected from Alzheimer disease, fronto-temporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, and progressive supranuclear palsy.
  • FDD fronto-temporal dementia
  • DLB dementia with Lewy bodies
  • Parkinson's disease vascular dementia
  • vascular cognitive impairment Huntington's disease
  • multiple sclerosis Creutzfeldt-Jakob disease
  • progressive supranuclear palsy progressive supranuclear palsy.
  • Embodiment 41 The method according to any one of Embodiments 35 to 38, wherein the agitation is associated with a neuropsychiatric condition.
  • Embodiment 42 The method according to Embodiment 41, wherein said neuropsychiatric condition is selected from schizophrenia, bipolar illness (such as mania, disorder), delirium, and depression.
  • said neuropsychiatric condition is selected from schizophrenia, bipolar illness (such as mania, disorder), delirium, and depression.
  • Embodiment 43 The method according to Embodiment 39, wherein the agitation is associated with sundown syndrome in dementia or Alzheimer's disease.
  • Embodiment 44 The method according to any one of Embodiments 35 to 38, wherein said agitation is associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures).
  • OPD/IPD procedure e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures.
  • Embodiment 45 The method according to any one of Embodiments 35 to 38, wherein said agitation is associated with an alcohol and substance abuse withdrawal.
  • Embodiment 46 The method according to any one of Embodiments 35 to 38, wherein said alpha-2 adrenergic agonist include, but is not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141
  • Embodiment 47 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein said agitation is associated with neurodegenerative diseases and said method comprises administering to said subject a therapeutically effective amount of Dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 48 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein said agitation is associated with sundowning syndrome in dementia or Alzheimer's disease and said method comprises: administering to said subject a therapeutically effective amount of Dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 49 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, intoxication and other neuropsychiatric conditions and said method comprises: administering to said subject a therapeutically effective amount of Dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 50 The method as embodied in embodiment 47, wherein said neurodegenerative disease is Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile x syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata.
  • said neurodegenerative disease is Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jak
  • Embodiment 51 The method as embodied in embodiment 50, wherein said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • Embodiment 52 The method as embodied in embodiments 47 to 49, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by oromucosal route including sublingual or buccal route in the form of pharmaceutical composition.
  • Embodiment 53 The method as embodied in embodiment 52, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by sublingual route.
  • Embodiment 54 The method as embodied in embodiments 47 to 49, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject (i.e. Human) is in a range of about 1 ⁇ g to about 105 ⁇ g, preferably 3 ⁇ g to 100 ⁇ g, more preferably 5 ⁇ g to 50 ⁇ g.
  • Embodiment 55 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with neurodegenerative diseases and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 56 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, post-traumatic stress disorder, alcohol and substance abuse withdrawal, intoxication and other neuropsychiatric conditions and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 57 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with sundowning syndrome in dementia or Alzheimers disease and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 58 A pharmaceutical composition for use in the treatment, prevention or reduction in the symptoms of agitation in a subject in need thereof wherein said chronic agitation is associated with neurodegenerative diseases and said pharmaceutical composition comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 59 A pharmaceutical composition for use in the treatment, prevention or reduction in the symptoms of agitation in a subject in need thereof wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, intoxication and other neuropsychiatric conditions and said pharmaceutical composition comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 60 A pharmaceutical composition for use in the treatment, prevention or reduction in the symptoms of agitation in a subject in need thereof wherein said agitation is associated with sundowning syndrome in dementia or Alzheimer's disease and said pharmaceutical composition comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 61 The method/composition as embodied in embodiments 55 and 58, wherein said neurodegenerative disease is selected from Frontotemporal dementia, Dementia, Alzheimer's disease, Parkinson's disease Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile x syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morpervan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata.
  • Frontotemporal dementia Dementia
  • Alzheimer's disease Parkinson's disease Multiple system atrophy
  • Embodiment 62 The method/composition as embodied in embodiment 61, wherein said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • Embodiment 63 The method/composition as embodied in embodiments 55 to 60, wherein said pharmaceutical composition is administered to said subject by an oromucosal route including sublingual or buccal route in the form of pharmaceutical composition.
  • Embodiment 64 The method/composition as embodied in embodiment 63, wherein said pharmaceutical composition is a tablet, film, gel etc.
  • Embodiment 65 The method/composition as embodied in embodiments 55 to 60, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject (i.e. Human) is in a range of about 1 ⁇ g to about 105 ⁇ g, preferably about 3 ⁇ g to 100 ⁇ g, more preferably about 5 ⁇ g to 50 ⁇ g and most preferably less than 10 ⁇ g.
  • Embodiment 66 Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof composition for treating a subject suffering from agitation associated with neurodegenerative disease.
  • Embodiment 67 Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof composition for treating a subject suffering from agitation associated with neuropsychiatric conditions including schizophrenia, bipolar disorder, bipolar mania, delirium, intoxication and other neuropsychiatric conditions.
  • Embodiment 68 Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof composition for treating a subject suffering from agitation associated with sundowning syndrome in dementia or Alzheimer's disease.
  • Embodiment 69 The use as embodied in embodiment 66, wherein neurodegenerative disease is selected from Frontotemporal dementia, dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile x syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata.
  • Neurodegenerative disease is selected from Frontotemporal dementia, dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob
  • Embodiment 70 The use as embodied in embodiment 69, wherein said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • Embodiment 71 A sublingual film composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with neurodegenerative disease.
  • Embodiment 72 A sublingual film composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with neuropsychiatric conditions such as bipolar disorder, schizophrenia. bipolar mania. Delirium, intoxication and other neuropsychiatric conditions.
  • neuropsychiatric conditions such as bipolar disorder, schizophrenia. bipolar mania. Delirium, intoxication and other neuropsychiatric conditions.
  • Embodiment 73 A sublingual film composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with sundowning syndrome in dementia or Alzheimer's disease.
  • Embodiment 74 A method of treating, preventing or reducing the symptoms of aggression in a subject in need thereof and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 75 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, the method comprises administering to said subject dexmedetomidine, or a pharmaceutically acceptable salt thereof through sublingual film in a dose range of 1 ⁇ g to 105 ⁇ g, preferably about 3 ⁇ g to 100 ⁇ g and more preferably about 5 ⁇ g to 50 ⁇ g, more preferably about 5 ⁇ g to 20 ⁇ g, most preferably less than 10 ⁇ g.
  • Embodiment 76 The method as embodied in embodiment 75, wherein the agitation is acute agitation.
  • Embodiment 77 The method as embodied in embodiment 75, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 78 The method as embodied in embodiment 77, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata , sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Frontotempo
  • Embodiment 79 The method as embodied in embodiment 77, wherein the neuropsychiatric conditions is selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal or intoxication.
  • Embodiment 80 The method embodied in embodiment 75, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to the said subject in a dose ranging from about 1 ⁇ g to about 105 ⁇ g; preferably from about 3 ⁇ g to about 100 ⁇ g, more preferably about 5 ⁇ g to about 50 ⁇ g, most preferably about 5 ⁇ g to about 20 ⁇ g.
  • Embodiment 81 A sublingual film comprising about 5 ⁇ g to about 100 ⁇ g of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation in a subject.
  • Embodiment 82 The sublingual film as embodied in embodiment 81, wherein the agitation is acute agitation.
  • Embodiment 83 The sublingual film as embodied in embodiment 82, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 84 The sublingual film as embodied in embodiment 83, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-LemliOpitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability, Agrypnia excitata , sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia,
  • Embodiment 85 The sublingual film as embodied in embodiment 83, wherein the neuropsychiatric conditions is selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal or intoxication.
  • Embodiment 86 The sublingual film as embodied in embodiment 81, wherein the film comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof ranging from about 1 ⁇ g to about 105 ⁇ g; preferably from about 3 ⁇ g to about 100 ⁇ g, more preferably about 5 ⁇ g to about 50 ⁇ g and most preferably about 5 ⁇ g to about 20 ⁇ g and less than 10 ⁇ g.
  • Embodiment 87 Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof for treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein the agitation or aggression is not perioperative agitation.
  • Embodiment 88 The use as embodied in embodiment 87, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 89 The use as embodied in embodiment 88, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-LemliOpitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability, Agrypnia excitata , sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Fronto
  • Embodiment 90 The use as embodied in embodiment 88, wherein the neuropsychiatric conditions are selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal and intoxication.
  • Embodiment 91 The use as embodied in embodiment 87, wherein the agitation is acute agitation.
  • Embodiment 92 The use as embodied in embodiment 87, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by oromucosal route preferably by sublingual or buccal route.
  • Embodiment 93 The use as embodied in embodiment 92, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered in form of pharmaceutical composition selected from the group comprising of tablet, film and gel.
  • Embodiment 94 The use as embodied in embodiment 87, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to the said subject in a dose ranging from about 1 ⁇ g to about 105 ⁇ g; preferably from about 3 ⁇ g to about 100 ⁇ g, more preferably about 5 ⁇ g to about 50 ⁇ g, most preferably about 5 ⁇ g to about 20 ⁇ g and most preferably less than 10 ⁇ g.
  • Embodiment 95 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof by sublingual route, wherein the agitation is not perioperative agitation.
  • Embodiment 96 A sublingual pharmaceutical composition for treating, preventing or reducing the symptoms of agitation in a subject in need thereof, the composition comprises a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and one or more pharmaceutical excipient, wherein the agitation is not perioperative agitation.
  • Embodiment 97 A sublingual use of dexmedetomidine, or a pharmaceutically acceptable salt thereof for treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein the agitation is not perioperative agitation.
  • Embodiment 98 A sublingual composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with neurodegenerative disease.
  • Embodiment 99 A sublingual composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with bipolar disorder, schizophrenia. bipolar mania. Delirium, intoxication and other neuropsychiatric conditions.
  • Embodiment 100 A sublingual composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with sundowning syndrome in dementia or Alzheimer's disease.
  • Embodiment 101 A method of treating, preventing or reducing the symptoms of agitation which is other than perioperative agitation in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof by sublingual route.
  • Embodiment 102 A sublingual film for treating, preventing or reducing the symptoms of agitation which is other than perioperative agitation in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 103 A method of treating, preventing or reducing the symptoms of aggression in a subject in need thereof and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 104 The method as embodied in embodiment 96, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 105 The method as embodied in embodiment 102, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-LemliOpitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability, Agrypnia excitata , sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Fronto
  • Embodiment 106 The method as embodied in embodiment 102, wherein the neuropsychiatric conditions is selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal or intoxication.
  • Embodiment 107 The method as embodied in embodiment 96, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by oromucosal route preferably by sublingual or buccal route.
  • Embodiment 108 The method as embodied in embodiment 105, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered in form of pharmaceutical composition selected from the group comprising of tablet, film or gel.
  • Embodiment 109 A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with other conditions such as OPD/IPD Procedures like MRI, CT or CAT Scan, Lumbar Puncture, Bone marrow aspiration/biopsy, Tooth extraction or other dental procedures and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • OPD/IPD Procedures like MRI, CT or CAT Scan, Lumbar Puncture, Bone marrow aspiration/biopsy, Tooth extraction or other dental procedures
  • said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 110 The methods/composition as embodied in the preceding embodiments, wherein the pharmaceutical composition includes intranasal formulations, parenteral formulations, oral formulations, transdermal formulations, liposomal formulations and the like.
  • Embodiments hereinabove are not intended to be limiting, and, in practicing the present invention, alternative or additional Embodiments may be provided.
  • composition for a typical Sub-lingual tablet formulation used for sublingual delivery Ingredients Quantity Ranges Dexmedetomidine HCl 50 (equivalent to base) micrograms Povidone 5.0 mg 1.0-10.0% Croscarmellose Sodium 7.0 mg 5-15% Sucralose 1.0 mg 0.05-3.0% Magnesium Stearate 0.75 mg 0.1-2.0% Talc 0.75 mg 0.1-2.0% Mannitol q.s 75.0 mg q.s. 100% Water q.s
  • Dexmedetomidine hydrochloride and excipients such as binder and sweetener are dissolved/dispersed into a pharmaceutically acceptable solvent (preferably water) and this solution is used to granulate the sifted blend of all other ingredients except lubricant and glidant in suitable mixer/granulator.
  • a pharmaceutically acceptable solvent preferably water
  • the granules are then dried in a fluid-bed drier or other suitable one such as tray drier.
  • the dried granules are then sized appropriately in quadro-co-mill or multi-mill.
  • the sized granules are then loaded into a suitable blender such as V-blender and lubricated with Magnesium stearate and Talc and then the final lubricated blend is then used for compressing into tablets of specific dimensions using appropriate tooling.
  • a suitable blender such as V-blender and lubricated with Magnesium stearate and Talc and then the final lubricated blend is then used for compressing into tablets of specific dimensions using appropriate tooling.
  • Dexmedetomidine hydrochloride along with film forming polymers and other excipients are dissolved/dispersed into a pharmaceutically acceptable solvent (preferably water) and the resulting solution is then coated (spread/cast) on an inert backing layer.
  • a pharmaceutically acceptable solvent preferably water
  • Dexmedetomidine hydrochloride containing polymeric layer is further dried, separated and cut into suitable sizes using appropriate die/tools and then packed as per the requirement.
  • Dexmedetomidine hydrochloride along with all other excipients are mixed in a suitable order.
  • the resulting solution/dispersion is then filled into spray canisters using appropriate tooling. They are further processed with Metered nozzles so that a specified amount of Dexmedetomidine is delivered after actuation each time.
  • Dexmedetomidine hydrochloride (Catalogue No. SML0956) was dissolved in Normal saline in order to yield the concentration of 1 mg/mi of the sublingual drops.
  • the resident-intruder model is an established preclinical model of aggression and agitation, and allows spontaneous and natural expression of both offensive aggression/agitation and defensive behavior in laboratory rodents in a semi natural laboratory setting.
  • rodents When rodents are exposed to a novel male in their home cage environment, they perceive the novel male animal as an “intruder” and demonstrate a repertoire of defensive behaviors such as ano-genital sniffing, chasing, biting and attacking (Nelson et al., ILAR Journal (2000) 41(3): 153-162).
  • Formulation tested The required quantity of Formulation 4 of dexmedetomidine hydrochloride was weighed and serial dilutions were made to obtain respective doses as per the Table 5. Dilutions were prepared fresh every day prior to dosing using 0.9% normal saline from the formulation 4 for the entire study.
  • each resident male rat was housed with a female rat for 8 days.
  • basal aggression in the resident males was tested by exposing them to an “intruder rat” for 10 minutes. Only animals that demonstrated aggression in this basal aggression test were used for the study. These animals were then randomized using body weight stratification method. The weight variation of the animals did not exceed 20% of the mean body weight in a group at the time of randomization. Animals were housed with the female rat for an additional day.
  • the resident animal was paired with intruder animal of an appropriate bodyweight such that the body weight of the resident was always higher than the intruder. This was to facilitate dominant, aggressive behavior in the resident animals.
  • animals were assigned a permanent number. Cages were identified by cage cards indicating the study number, study code, group number, sex, dose, cage number and animal number details.
  • the behavior of the resident rat was recorded using an overhead video camera for 15 minutes and offline behavioral analysis was done using the Noldus Ethovision XT software. To distinguish the resident rat from the intruder rat in the video recording, the intruder rat was marked with non-toxic paint.
  • we quantified various behavioral parameters such as anogenital sniffing, chasing, biting, attacking and latency to attack as well as neutral behavioral parameters such as exploration grooming, and immobile quiet time.
  • the rats demonstrate a variety of defensive agitated behaviors such as anogenital sniffing, chasing, biting and attacking (indices of agitative and aggressive behavior) when exposed to a novel male in their home cage environment.
  • the non-resident male is perceived as intruder and the resident male gets agitated and attacks the intruder male to protect their home territory.
  • vehicle treated rats demonstrated a wide repertoire of aggressive behaviors and the intruder rat was subjected to anogenital sniffing, attack, chasing and biting by the resident or dominant rat.
  • Dexmedetomidine hydrochloride (Dex) administered sublingually reduced the frequency and duration of these behaviors in a dose related manner ( FIG. 1 A , and FIG. 1 B ). Significant reduction was observed in chasing and attacking compared to vehicle control group. Similarly, intravenous administration of dexmedetomidine hydrochloride (Dex) reduced all the indices of aggressive and agitated behaviors ( FIG. 1 C and FIG. 1 D ). A significant reduction in anogenital sniffing, biting and attacking compared to vehicle controls was observed at doses above 0.5 ⁇ g/kg ( FIG. 1 C and FIG. 1 D ). Reference compound diazepam (3 mg/kg, i.p) also produced significant reduction in all the indices of aggressive and agitated behaviors evaluated in this study ( FIG. 1 A- 1 D ).
  • the resident-intruder model is an established preclinical model of aggression/agitation and allows spontaneous and natural expression of both offensive aggression/agitation and defensive behavior in laboratory rodents in a semi natural laboratory setting.
  • Sublingual administration of Dexmedetomidine hydrochloride resulted in a dose related reduction in several behavioral indices of aggression and agitation such as anogenital sniffing, chasing, attacking and biting.
  • doses of 1-1.5 ⁇ g/kg doses administered sublingually or intravenously effectively reduced the behavioral indices of aggression and agitation without majorly impacting the neutral behaviors.
  • Dexmedetomidine hydrochloride effectively reduces various indices of agitation and aggression in rat resident intruder model. Dose of 1-1.5 ⁇ g/kg effectively reduced the behavioral indices of aggression and agitation without majorly impacting the neutral behaviors. In the present study the efficacy of sublingually administered Dexmedetomidine hydrochloride correlates with intravenously administered Dexmedetomidine hydrochloride at these doses (Table 6).
  • the human equivalent sublingual doses are calculated to be 0.161 ⁇ g/kg & 0.242 ⁇ g/kg.
  • the total human equivalent dose for a 60-kg human would be 10 and 15 ⁇ g (https://www.fda.gov/downloads/drugs/guidances/ucm078932.pdf).
  • Example 3 Estimation of Dexmedetomidine (0.5-3 ⁇ g/Kg) in Rat Plasma Samples by LC-MS/MS
  • a standard stock solution of dexmedetomidine hydrochloride was prepared by dissolving 1.358 mg of dexmedetomidine hydrochloride in 1358 ⁇ l of milli-Q water to achieve a concentration of 829.071 mg/ml.
  • Working solutions of different concentrations were prepared by using diluent (methanol:water (50:50) % v/v).
  • Tolbutamide was used as an internal standard and its stock solution was prepared by dissolving 25 mg of tolbutamide in 1000 ⁇ l of DMSO to achieve a concentration of 25 mg/ml.
  • Working solutions of different concentrations were prepared by using a diluent (acetonitrile: water (50:50) % v/v).
  • Solution preparation for SPE and chromatography Mobile phase A (10 mm ammonium formate, pH 3.50): 0.6306 gms of ammonium formate was weighed and transferred to a 1000 ml reagent bottle. To this, 1000 ml of milli q water was added and pH of the resulting solution was adjusted to 3.5 using formic acid.
  • Diluent methanol:water (50:50) % v/v: 50 ml of methanol was mixed with 50 ml of milli-q water. Resulting solution was used as diluent.
  • Wash solution 100 ⁇ l of ammonia was mixed with 100 ml of milli q. Resulting solution was used as wash solution.
  • Elution solvent 100 ⁇ l of formic acid was mixed with 100 ml of acetonitrile. Resulting solution was used as elution solvent.
  • WCX SPE 96 well plate was used for sample preparation. 50 ⁇ l of plasma sample was used for extraction. Along with study samples, one set of linearity and two sets of quality controls (QC) were also processed.
  • Sample pretreatment To 50 ⁇ l of plasma, 10 ⁇ l of tolbutamide working solution was added (Tolbutamide 250 ng/ml). After mixing, 50 ⁇ L of buffer solution (10 mM Ammonium Formate pH 3.5) was added. Contents were vortex mixed and loaded to preconditioned SPE plate.
  • the cartridges After placing the cartridges in the negative pressure SPE unit, they were conditioned by passing 200 ⁇ l of 100% methanol followed by 200 ⁇ l of water. The pretreated plasma samples were then loaded to the pre-conditioned cartridges.
  • Mean plasma concentrations of Dexmedetomidine in various rat plasma samples at various time points was determined by LC-MS/MS method using Analyst 1.6.2 software (Table 8 and FIGS. 4 A and 4 B ) with a calibration curve in the range of 0.011-53.061 ng/ml prepared in blank rat plasma matrix. The calibration curve was fitted by linear regression. The concentrations in the QC and test samples (pg/mL) were obtained from the Analyst software based on the calibration curve.
  • Acceptance criteria for the calibration curve and QCs are as follows: 1) At least 75% of the non-zero calibration standards must be included in the calibration curve with all back-calculated concentrations within ⁇ 20% deviation from nominal concentrations (except for the lower level of quantification, LLOQ, where ⁇ 20% deviation is acceptable). 2) The correlation coefficient (r) of the calibration curve must be greater than or equal to 0.99. 3) At least two-thirds (4 out of 6) QC samples must be within ⁇ 20% relative error (accuracy)
  • HCl 7.2 14.2 4.9 3.5 0.5 ⁇ g/kg, 0.5 ⁇ g/kg, SL i.v. II- BLQ 51 ⁇ 47 ⁇ 43 ⁇ 13 ⁇ 19 ⁇ VI- BLQ 174 ⁇ 90 ⁇ 45 ⁇ 63 ⁇ BLQ Dex. HCl 44.7 22.4 13.5 2.8 7.07 Dex. HCl 12.5 12.1 1.7 58.0 1 ⁇ g/kg, 1 ⁇ g/kg, SL i.v. III- BLQ 84 ⁇ 27 ⁇ 31 ⁇ 37 ⁇ 36 ⁇ VII- BLQ 158 ⁇ 114 ⁇ 65 ⁇ 31 ⁇ 21 ⁇ Dex. HCl 37.7 7.1 5.5 16.3 6.36 Dex.
  • Plasma concentrations following administration of dose of 1 ⁇ g/kg (via sublingual and intravenous route) between 15 to 30 min range from 43 ⁇ 13.5 to 90 ⁇ 12.1 ⁇ g/ml (Table 8).
  • plasma concentrations following administration of dose of 1.5 ⁇ g/kg (via sublingual and intravenous route) between 15 to 30 min range from 27 ⁇ 7.1-114 ⁇ 1.7 pg/ml (Table 8).

Abstract

The present invention discloses a method of treating agitation or the signs of agitation in a subject comprising the sublingual administration of an effective amount of an alpha-2 adrenergic agonist, more particularly Dexmedetomidine, or a pharmaceutically acceptable salt thereof. The method is particularly suitable for the treatment of agitation associated with neurodegenerative and/or neuropsychiatric diseases. The present invention also discloses the sublingual administration of an alpha-2 adrenergic agonist, more particularly Dexmedetomidine or a pharmaceutically acceptable salt thereof at a dose that is effective to treat agitation or the signs of agitation in a subject, but does not cause significant sedation.

Description

    CROSS REFERENCE TO RELATED APPLICATION
  • This application is a continuation of U.S. patent application Ser. No. 16/474,882, filed Jun. 28, 2019, which is a U.S. national stage application of International Patent Application No. PCT/US2017/069030, filed Dec. 29, 2017, which claims the benefit of priority to U.S. Provisional Application No. 62/441,164 filed Dec. 31, 2016, U.S. Provisional Application No. 62/471,393 filed Mar. 15, 2017 and U.S. Provisional Application No. 62/542,323 filed Aug. 8, 2017, the disclosures of which are herein incorporated by reference in their entirety for all purposes.
  • FIELD OF THE INVENTION
  • The present invention discloses a method of treating agitation or the signs of agitation in a subject comprising sublingually administering an effective amount of an alpha-2 adrenergic agonist, more particularly Dexmedetomidine or a pharmaceutically acceptable salt thereof. The present invention also discloses a sublingual composition for treating agitation or the signs of agitation comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients, along with the preparation thereof.
  • BACKGROUND OF THE INVENTION
  • Agitation is an umbrella term that can refer to a range of behavioral disturbances or disorders, including aggression, combativeness, hyperactivity, and disinhibition. Agitation is a nonspecific constellation of relatively unrelated behaviors that can be seen in several different clinical conditions, usually presenting a fluctuating course. Agitation may be caused by several different medical conditions and drug interactions or by any circumstances that worsen the person's ability to think. Multiple underlying pathophysiologic abnormalities are mediated by dysregulations of dopaminergic, serotonergic, noradrenergic, and GABAergic systems. Agitation is characterized by non-productive, diffuse and excessive over-activity both motor (akathisia) and cognitive, and accompanied by an inner unpleasant tension. The key to safety is to intervene early to prevent progression of agitation to aggression and violence.
  • Agitation can be associated with neurodegenerative disorders. One of the important manifestations of long-term progressive neurodegenerative process is clinically known as dementia. Dementias include Alzheimer's disease dementia (AD), Fronto-temporal dementia (FTD), Vascular dementia, Lewy body disease (LBD), and Down dementia. Dementia in adults, gradually destroy a person's memory and ability to learn, reason, make judgments, communicate and carry out daily activities. In later stages, patients may experience changes in personality and behavior, such as anxiety, suspicion, agitation and aggression.
  • Sebastiaan Engelborghs et al., in Neurochemistry International 2007 November, 52(6): 1052-60, disclosed that, in frontotemporal dementia, increased activity of dopaminergic neurotransmission and altered serotonergic modulation of dopaminergic neurotransmission are associated with agitated and aggressive behavior respectively. Pia Jul et al., in Journal of Alzheimer's disease 2015 September, 49(3):783-95, disclosed that rTg4510 mice exhibited P301L-tau-dependent hyperactivity, and agitation-like phenotypes in these mice may form a correlation to some of the behavioral disturbances observed in advanced Alzheimer's disease (AD) and Frontotemporal dementia (FTD). Nathan Hermann et. al., in Journal of Neuropsychiatry 2004 August, 16(3): 261-276, disclosed that a compensatory increase in activity within the noradrenergic system may contribute to the behavioral and psychological symptoms of agitation and aggression in Alzheimer's disease.
  • Agitation can also be associated with neuropsychiatric conditions such as schizophrenia, bipolar illness such as bipolar disorder or mania, depression, delirium, etc or agitation can be associated with alcohol and substance abuse withdrawal. Acute agitation, represented by a state of motor restlessness and accompanying mental tension, is a serious medical problem that can be present in some psychiatric disorders, including schizophrenia and bipolar mania, and may escalate quickly to aggressive behavior. Acute agitation is characterized by signs that include pacing, hand wringing, fist clenching, pressured speech, yelling, and threatening people with escalated agitation.
  • To date, there is no single medication considered as the “standard of care” for treating agitation in patients with dementia or schizophrenia. Generally, three classes of medications are used most frequently, depending on the severity of the agitation, namely first-generation antipsychotics, second-generation antipsychotics, and benzodiazepines, administered orally, intramuscularly or intravenously. Intramuscular injection of typical antipsychotics and benzodiazepines, given alone or in combination, has been a treatment of choice for agitation over the past few decades. The currently preferred treatment paradigm for acute agitation is to use atypical antipsychotic drugs administered with or without supplemental benzodiazepines.
  • More specifically, patients with agitation are usually prescribed beta blockers such as propranolol and Pindolol, anxiety medications such as Buspirone, benzodiazepines such as Lorazepam, anti-convulsants such as Valproate and Lamotrigine, anti-psychotics such as Haloperidol, Droperidol, Ziprasidone and other high-potency dopamine-blocking agents, and atypical antipsychotics such as Olanzapine. However, Buspirone, Valproate, Haloperidol, Droperidol and Ziprasidone have potential adverse effects, and optimal dosage and long-term efficacy in the management of chronic agitation in dementia is very limited. Lorazepam is only effective for treating agitation in patients when used before medical procedures. Loxapine (an antipsychotic) is FDA approved for treating agitated patients via inhalation, but is associated with a black box warning for bronchospasm and increased mortality in elderly patients with dementia-related psychosis (FDA label, Loxapine or Adasuve®). Olanzapine, Ziprasidone or its combination with Haloperidol, is also associated with QT prolongation, and extrapyramidal side effects should be watched very carefully in hospital set ups. Reports of adverse events (including eight fatalities) associated with intramuscular olanzapine underscores the need to follow strict prescribing guidelines and avoid simultaneous use with other CNS depressants.
  • The Expert Consensus Guidelines for treatment of behavioral emergencies cite speed of onset as one of the most important factors in choosing a drug and its route of administration. However, antipsychotic medications can take from days to weeks before having a robust antipsychotic effect. Nevertheless, they do generally have a calming effect on agitated patients within minutes. For example, benzodiazepines or fast-acting sedatives quickly calm a severely agitated patient, but continuous treatment with these drugs leads to tolerance.
  • Therefore, the treatment of agitation in patients with neuropsychiatric conditions (such as schizophrenia or bipolar mania) and neurodegenerative diseases is still limited because of the potential for significant side effects associated with currently used drugs, their route of administration (intravenous/intramuscular) and the consequent need for hospital set ups for administering these drugs. In an ideal situation, an anti-agitation drug for schizophrenics or dementia patient should have a rapid onset of calming without sedation, be well tolerated and easy to administer with a high safety margin.
  • Alpha-2 adrenergic agonists have been used therapeutically for a number of conditions, including hypertension, congestive heart failure, angina pectoris, spasticity, glaucoma, diarrhea and for suppression of opiate withdrawal symptoms. Examples of alpha-2 adrenergic agonists include Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-aminomethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione, and 4-(3-hydroxymethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione. The inventors of the present invention have unexpectedly found that the sub-lingual administration of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof is a particularly effective and safe intervention for the treatment of agitation.
  • (S)-4-[1-(2,3-Dimethylphenyl)ethyl]-3H-imidazole (Dexmedetomidine) is commercially available as an injectable formulation for sedation of initially intubated and mechanically ventilated patients during treatment in an intensive care setting, and for non-intubated patients prior to and/or during surgical and other procedures.
  • Dexmedetomidine is reported to have anti-agitational effects when administered intravenously or buccally during surgical procedures and intensive care unit (ICU) setups. For example, Ibacache et. al., in Anesthesia & Analgesia 2004 January; 98(1):60-3, discloses the administration of an intravenous single-dose of Dexmedetomidine to reduce agitation following sevoflurane anesthesia in children. Other intravenous administrations are reported by Jeanne Boyer et al., in Nursing Critical care 2010 January, 5(1):30-34, Yahya Shehabi et. al., in Anesthetic Intensive Care 2010 January, 38(1):82-90, and Joseph D. Tobias in Journal of Pediatric Pharmacology Therapeutic, January-March 2010, 15(1): 43-48. NCT 02720705 (clinical trial identification number from clinicaltrials.gov) discloses the administration of transbuccal Dexmedetomidine for the prevention of emergence agitation in pre-school children treated with sevoflurane in an intensive care unit setting.
  • The sublingual use of Dexmedetomidine is disclosed in WO 2016/061413. However, the focus of WO 2016/061413 is the administration of Dexmedetomidine sublingually at doses appropriate to treat sleep disorders and induce significant sedation. We have now surprisingly found that Dexmedetomidine or a pharmaceutically acceptable salt thereof, administered sublingually, can effectively treat agitation, including agitation associated with neurodegenerative diseases (e.g. Alzheimer's disease, fronto-temporal dementia, and sundown syndrome in Alzheimer's disease/dementia), agitation associated with neuropsychiatric conditions (e.g. bipolar disorder, schizophrenia, bipolar mania, delirium and depression), agitation associated with alcohol and substance abuse withdrawal or agitation associated with other conditions such as OPD/IPD procedures (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures). The dose to be administered sublingually may be selected to be effective to treat agitation, yet insufficient to causing significant sedation.
  • SUMMARY OF THE INVENTION
  • The present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof sublingually to the subject, wherein the said agitation is associated with a neurodegenerative disease like dementia, Alzheimer's disease, frontotemporal dementia, or Parkinsonism, or associated with a neuropsychiatric condition like schizophrenia, bipolar disorder, bipolar mania, delirium, or depression, or associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), or associated with an alcohol and substance abuse withdrawal. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • In a preferred aspect, the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • Another aspect of the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with neurodegenerative disease, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • Yet another object of the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with dementia, Alzheimer's disease, frontotemporal dementia, Parkinsonism or other neurodegenerative diseases, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • Another object of the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, depression, or another related neuropsychiatric condition, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • A further object of the present invention provides a method of treating, preventing or reducing the signs of agitation in a subject in need thereof, wherein said agitation is associated with sundown syndrome in Alzheimer's disease/dementia, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • Yet another objective of the present invention provides a method for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an OPD/IPD procedure (e.g., MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • Yet another objective of the present invention provides a method for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an alcohol and substance abuse withdrawal, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • A further aspect of the present invention provides a sublingual composition for treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with a neurodegenerative disease, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutical acceptable carriers and/or excipients.
  • Another aspect of the present invention provides a sublingual composition for treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, depression, or another related neuropsychiatric condition, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically acceptable carriers and/or excipients.
  • An additional aspect of the present invention provides a sublingual composition for treating agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with sundown syndrome in Alzheimer's disease/dementia, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients.
  • Yet another aspect of the present invention provides a sublingual composition for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients.
  • Yet another aspect of the present invention provides a sublingual composition for treating agitation or the signs associated with agitation in a subject in need thereof, wherein said agitation is associated with an alcohol and substance abuse withdrawal, and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients.
  • Another object of the present invention provides a sublingual composition comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients, wherein said sublingual composition is selected from the group consisting of a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • A further object of the present invention provides a method of sublingually administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof to a subject's oral mucosa to treat agitation or the signs of agitation at a dosage which does not cause significant sedation.
  • In a particular aspect of the invention, the dosage administered sublingually may conveniently be in the range of between about 3 micrograms to about 100 micrograms, Examples of suitable dosages include: about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms. The dose may be administered one or more times a day.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A. Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on cumulative duration of aggressive and agitated behaviors. Data expressed as Mean f SEM. One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 1B. Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on frequency of aggressive and agitated behaviors. Data expressed as Mean±SEM. One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 1C. Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on cumulative duration of aggressive and agitated behaviors. Data expressed as Mean±SEM. One-way ANOVA followed by Dunnett's post-hoc test. **p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 1D. Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on frequency of aggressive and agitated behaviors. Data expressed as Mean±SEM. One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 2A. Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Latency to attack. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 2B. Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Latency to attack. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 3A. Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Cumulative duration of Neutral behaviors such as grooming, and exploration. Data expressed as Mean f SEM. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 3B. Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Frequency of Neutral behaviors such as grooming, and exploration. Data expressed as Mean f SEM. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 3C. Effect of sublingually administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Neutral behaviors such as immobile/quiet time. Data expressed as Mean f SEM. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 3D. Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Cumulative duration of Neutral behaviors such as grooming, and exploration. Data expressed as Mean t SEM. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 3E. Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Frequency of Neutral behaviors such as grooming, and exploration. Data expressed as Mean±SEM. Data is expressed as Mean t SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 3F. Effect of intravenously administered Dexmedetomidine hydrochloride (Dex) at varying doses (0.5-3 μg/kg) on Neutral behaviors such as immobile/quiet time. Data expressed as Mean±SEM. Data is expressed as Mean±SEM. Statistical analysis was performed by One-way ANOVA followed by Dunnett's post-hoc test. *p<0.05 **p<0.01, ***p<0.001 and ****p<0.0001 vs vehicle controls (vehicle).
  • FIG. 4A: Mean plasma concentrations following Sublingual (SL) Dexmedetomidine hydrochloride administration in rats. Data expressed as Mean±SD
  • FIG. 4B: Mean plasma concentrations following Intravenous (IV) Dexmedetomidine hydrochloride administration in rats. Data expressed as Mean±SD
  • DETAILED DESCRIPTION OF THE INVENTION I. Abbreviations
  • The following abbreviations are used throughout this specification:
  • AD: Alzheimer's disease
    AUC: Area under the curve
  • BZDs: Benzodiazepines
  • CNS: Central nervous system
    CT/CAT scan: computed tomography scan
    Cmax: Maximum (or peak) serum concentration that a drug achieves in a specified compartment
    EPS: Extrapyramidal side effects
  • FD & C: Federal Food, Drug, and Cosmetic
  • FTD: Fronto-temporal dementia
  • GABA: Gamma-aminoautyric Acid 5-HT: 5-Hydroxytryptamine
  • ICU: Intensive care unit
    IPD: In-Patient department
    MRI: Magnetic resonance imaging
  • Mg: Milligram NE: Nor-epinephrine
  • OPD: Out-patient department
    PTSD: Post-traumatic stress disorders
    RSS: Ramsay sedation score
    RIT: Rat intruder test
    SLOS: Smith-Lemli Opitz syndrome
    Tmax: Time at which the Cmax is observed.
  • II. Definitions
  • It will be understood that the terminology used herein is for the purpose of describing embodiments only, and is not intended to be limiting. As used in this specification, the singular forms “a”, “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a solvent” includes one or more such solvents and the like.
  • 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 the invention pertains. Although other methods and materials similar, or equivalent, to those described herein can be used in the practice of the present invention, the preferred materials and methods are described herein.
  • The terms “treating,” and “treatment,” as used herein refer to curative therapy, prophylactic therapy, and/or preventative therapy and can be used interchangeably.
  • As used herein, unless indicated otherwise, the terms “pharmaceutical composition”, “composition”, “formulation” and “composition of the invention,” are used interchangeably. Unless stated otherwise, the terms are meant to encompass, and are not limited to, pharmaceutical compositions containing drug substance i.e. Dexmedetomidine. The composition may also contain one or more “excipients” that are “inactive ingredients” or “compounds” devoid of pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease or to affect the structure or any function of the human body.
  • As used herein, the term “an effective amount” is interchangeable with “therapeutically effective dose,” or “therapeutically effective amount,” and refers to an amount sufficient to produce the desired effect. An effective amount is sufficient to cause an improvement in a clinically significant condition of the subject.
  • As used herein, “pharmaceutically acceptable salt” refers to a salt known to be non-toxic and commonly used in the pharmaceutical literature. Typical inorganic acids used to form such salt include hydrochloric, hydrobromic, hydroiodic, nitric, sulfuric, phosphoric, hypophosphoric, and the like. Salts derived from organic acids, such as aliphatic mono and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic and hydroxyl alkandioic acids, aromatic acids, aliphatic and aromatic sulfonic acids may also be used. A preferred salt is the hydrochloride salt.
  • As used herein, the term “subject” preferably refers to a human patient. In some embodiments, the subject can be any animal, including non-human mammals, such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates.
  • The term “agitation”, as used herein, means irritability, emotional outburst, impaired thinking, or excess motor and verbal activity that may occur due to either dysfunction of specific brain regions such as frontal lobes or due to dysfunction of neurotransmitter systems such as dopamine and nor-epinephrine. In the present invention, agitation also includes aggression and hyper-arousal in post-traumatic stress disorder. The agitation may be acute or chronic.
  • The term “the signs of agitation” includes excessive motor activity (examples include: pacing, rocking, gesturing, pointing fingers, restlessness, performing repetitious mannerisms), verbal aggression (e.g. yelling, speaking in an excessively loud voice, using profanity, screaming, shouting, threatening other people), physical aggression (e.g. grabbing, shoving, pushing, clenching hands into fists, resisting, hitting others, kicking objects or people, scratching, biting, throwing objects, hitting self, slamming doors, tearing things, and destroying property).
  • The term “acute agitation” means agitation that occurs rapidly and is severe and sudden in onset. Acute agitation may be associated with, for example, neurodegenerative disease and neuropsychiatric conditions, although it may particularly exist in neuropsychiatric conditions. Acute agitation may lead to chronic agitation if it remains untreated.
  • The term “chronic agitation” means agitation developed over a long period of time, and is less severe than acute agitation. Chronic agitation may be associated with, for example, neurodegenerative disease and neuropsychiatric conditions, although it may particularly exist in neurodegenerative diseases.
  • The term “neurodegenerative disease” includes, but is not limited to, Alzheimer disease, frontotemporal dementia (or Pick's disease), Dementia, Dementia with Lewy bodies, post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, progressive supranuclear palsy or other related neurodegenerative diseases.
  • The term “neuropsychiatric conditions” includes, but is not limited to, schizophrenia, bipolar illness (bipolar disorder, bipolar mania), depression, delirium or other related neuropsychiatric conditions.
  • “Sundown syndrome” is a late-day circadian syndrome of increased confusion and restlessness, generally in a patient with some form of dementia. It seems to occur more frequently during the middle stages of Alzheimer dementia. It seems to subside with the progression of a patient's dementia. About 20-45% of Alzheimer type patients will experience some sort of sundowning confusion. Confusion and agitation worsen in the late afternoon and evening, or as the sun goes down.
  • The term “perioperative agitation” means agitation before, during or after any surgical procedure or ICU agitation unassociated with a neurodegenerative disease or neuropsychiatric condition.
  • The term “sublingual” literally means “under the tongue” and refers to a method of administering substances via the mouth in such a way that the substances are rapidly absorbed via the blood vessels under the tongue rather than via the digestive tract. Sublingual absorption occurs through the highly vascularized sublingual mucosa, which allows a substance direct access to the blood circulation, thereby providing for direct systemic administration independent of gastrointestinal influences and avoiding undesirable first-pass hepatic metabolism. Accordingly, the total amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof in the formulation may be reduced, thereby reducing the likelihood of deleterious side effects and providing a cost benefit to the manufacturer.
  • “Sedation” as used herein means depressed consciousness in which a patient or subject retains the ability to independently and continuously maintain an open airway and a regular breathing pattern, and to respond appropriately and rationally to physical stimulation and verbal commands. As used herein “without causing significant sedation” means that the patient experiences a level of sedation not greater than Level 3 on the Ramsay Sedation Scale. Level 3 means sedated but responds to commands.
  • III. Methods
  • The present invention provides a method of treating agitation or the signs of agitation in a subject comprising administering an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • In one embodiment, the alpha-2 adrenergic agonist includes, but is not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-aminomethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione, and 4-(3-hydroxymethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione or a pharmaceutically acceptable salt thereof.
  • In one preferred embodiment, the present invention provides a method of treating agitation or the signs of agitation in a subject comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject. In a particular aspect, the agitation is suppressed without also causing significant sedation.
  • Agitation may be effectively treated using a relatively low dose of Dexmedetomidine or a pharmaceutically acceptable salt thereof via the sublingual route. Consequently, in addition to providing relief from agitation without causing significant sedation, the treatment is also effective with reduced or no side effects (for example, cardiac or respiratory side effects).
  • In a further embodiment, the present invention is directed to a method of treating agitation or the signs of agitation in a subject comprising administering Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to the subject to provide fast-acting relief without a substantial portion of Dexmedetomidine or its pharmaceutically acceptable salt thereof passing into the liver of the patient.
  • In another embodiment, the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof via a sublingual composition to the subject, wherein the sublingual composition is selected from a film, wafer, patch, lozenge, gel, spray, tablet, and liquid drops.
  • In a further embodiment, the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to the subject an effective amount of an alpha-2 adrenergic agonist together with one or more pharmaceutically acceptable carriers and/or excipients via a sublingual composition, wherein the sublingual composition is a sublingual film. In a particular aspect, the agitation is associated with a neurodegenerative disease or neuropsychiatric condition. In another particular aspect, the treatment is effective without causing significant sedation.
  • In a further embodiment, the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to the subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers and/or excipients via a sublingual composition, wherein the sublingual composition is a sublingual film. In a particular aspect, the agitation is associated with a neurodegenerative disease or neuropsychiatric condition. In another particular aspect, the treatment is effective without causing significant sedation.
  • In yet other embodiment, the present invention provides a method of treating agitation or signs of agitation in a subject in need thereof, comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof at a dosage that does not cause a significant sedation. Suitable alpha-2 adrenergic agonists include, but are not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-aminomethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione, and 4-(3-hydroxymethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione or a pharmaceutically acceptable salt thereof. In a particular aspect of the invention, the dosage of alpha-2 adrenergic agonist used in the composition is from about 3 micrograms to about 100 micrograms.
  • In another embodiment, the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually at a dosage that does not cause significant sedation. In a particular aspect of the invention, the dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof used in the sublingual composition is from about 3 micrograms to about 100 micrograms. Examples of suitable dosages include: about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms. The dose may be administered one or more times a day.
  • In a further embodiment, the present invention provides a method of treating agitation or the signs of agitation in a subject in need thereof, comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually at a dosage of from about 0.05 micrograms/kg weight of subject to about 1.5 micrograms/kg weight of subject. Examples of suitable dosages include: about 0.1 micrograms/kg to about 1 micrograms/kg, about 0.1 micrograms/kg to about 0.5 micrograms/kg, about 0.1 micrograms/kg to about 0.4 micrograms/kg, about 0.1 micrograms/kg to about 0.3 micrograms/kg, about 0.1 micrograms/kg to about 0.2 micrograms/kg, about 0.07 micrograms/kg, about 0.05 micrograms/kg, about 0.1 micrograms/kg, about 0.2 micrograms/kg, about 0.3 micrograms/kg, about 0.4 micrograms/kg, about 0.5 micrograms/kg, about 0.6 micrograms/kg, about 0.7 micrograms/kg, about 0.8 micrograms/kg, about 0.9 micrograms/kg, about 1.0 micrograms/kg, about 1.1 micrograms/kg, about 1.2 micrograms/kg, about 1.3 micrograms/kg, about 1.4 micrograms/kg, about 1.5 micrograms/kg. The dose may be administered one or more times a day.
  • In yet other embodiment, the present invention provides a method of treating agitation or signs of agitation associated with neurodegenerative disease in a subject in need thereof, comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof at a dosage that does not cause a significant sedation. Suitable alpha-2 adrenergic agonists include, but are not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-aminomethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione, and 4-(3-hydroxymethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione or a pharmaceutically acceptable salt thereof. The dosage of alpha-2 adrenergic agonist used in the composition is conveniently from about 3 micrograms to about 100 micrograms.
  • In a yet further embodiment, the present invention provides a method of treating agitation or the signs of agitation associated with neurodegenerative disease in a subject in need thereof, comprising sublingually administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof at a dosage that does not cause unwanted (e.g., significant) sedation. The dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof used may conveniently be from about 3 micrograms to about 100 micrograms, e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms, about 5 micrograms, about 6 micrograms, about 7 micrograms, about 8 micrograms, about 9 micrograms, about 10 micrograms, about 12 micrograms, about 14 micrograms, about 16 micrograms, about 18 micrograms. The dose may be administered one or more times a day.
  • In yet other embodiment, the present invention provides a method of treating agitation or signs of agitation associated with neuropsychiatric condition in a subject in need thereof, comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof at a dosage that does not cause a significant sedation. Suitable alpha-2 adrenergic agonists include, but are not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine, Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-aminomethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione, and 4-(3-hydroxymethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione or a pharmaceutically acceptable salt thereof. The dosage of alpha-2 adrenergic agonist used in the composition is conveniently from about 3 micrograms to about 100 micrograms.
  • In another embodiment, the present invention provides a method of treating agitation or the signs of agitation associated with neuropsychiatric condition in a subject in need thereof, comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually at a dosage that does not cause significant sedation. The dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof used in a sublingual composition may conveniently be from about 3 micrograms to about 100 micrograms, e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms, about 5 micrograms, about 6 micrograms, about 7 micrograms, about 8 micrograms, about 9 micrograms, about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms. The dose may be administered one or more times a day.
  • The level of acceptable sedation when treating a subject according to a method of the present invention is preferably at or below Level 3 according to the Ramsay sedation scoring (RSS) system. Thus, a particular embodiment of the present invention provides a method of treating agitation or the signs of agitation in a human subject in need thereof, comprising administering Dexmedetomidine or a pharmaceutically acceptable salt thereof sublingually to said subject at a dose in the range of about 3 micrograms to about 100 micrograms, thereby achieving an RSS at or below Level 3 (e.g. Level 2 or Level 3).
  • IV. Pharmaceutical Compositions
  • The present invention also provides sublingual pharmaceutical compositions comprising an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof, preferably Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • The sublingual pharmaceutical compositions of the present invention may also comprise a pharmaceutically acceptable carrier and/or excipient. Suitable pharmaceutically acceptable carriers include water, sodium chloride, binders, penetration enhancers, diluents, lubricants, flavouring agents, coloring agents and so on.
  • The sublingual pharmaceutical compositions of the present invention may be administered to a subject alone or in combination with one or more other suitable active ingredients.
  • In one embodiment, the present invention provides a sublingual pharmaceutical composition comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof for the treatment of agitation in a subject, e.g., agitation associated with neurodegenerative disease, sundown syndrome in Alzheimer's disease or dementia. In a particular aspect, the sublingual pharmaceutical composition effectively treats agitation in a subject without causing significant sedation.
  • In another embodiment, the present invention provides a sublingual pharmaceutical composition comprising an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof for the treatment of agitation in a subject associated with schizophrenia, bipolar disorder, bipolar mania, other bipolar illness, depression, delirium or another related neuropsychiatric condition. In a particular aspect, the sublingual pharmaceutical composition effectively treats agitation in a subject without causing significant sedation.
  • The sublingual pharmaceutical composition of the present invention may be, for example, a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • In one embodiment of the invention, the sublingual pharmaceutical composition is in the form of a tablet or packed powder.
  • In another embodiment of the invention, the sublingual pharmaceutical composition is in the form of a patch or film (e.g. thin film). The patch may have adhesive qualities to prevent movement or swallowing of the patch. The patch may be ingestible in case of accidental swallowing or to allow for its easy disposal, or the patch may be removed from under the tongue after a prescribed time.
  • In yet another embodiment of the invention, the sublingual pharmaceutical composition is in the form of a paste, gel or ointment. The viscosity of the paste, gel or ointment can be adjusted to allow for retention under the tongue.
  • In a further embodiment of the invention, the sublingual pharmaceutical composition is in a liquid (e.g. as a solution, suspension or emulsion), and may be, for example, presented as a spray or as drops. Solutions include the active ingredient together with a diluent such as water, normal saline, sodium chloride solution, or any other suitable solvent such as propylene glycol, glycerol, ethyl alcohol and so on. The diluent for the solution may particularly be physiological saline solution or water. The amount of solution administered may conveniently be about 0.01 ml to about 1 ml (e.g., about 0.025-0.5 ml).
  • The non-solid compositions of the invention may conveniently be administered by spraying, dripping, painting or squirting the composition under the tongue.
  • In a particular embodiment of the invention, Dexmedetomidine or a pharmaceutically acceptable salt thereof is sublingually administered in liquid form, e.g. in a flavored or unflavored physiological saline solution. The liquid composition may conveniently be administered under the tongue as drops or as a spray.
  • Dexmedetomidine, or a pharmaceutically acceptable salt thereof may conveniently represent from about 0.001% to about 99.99% of the overall composition, e.g. about 0.01% to about 90%, more particularly about 0.01% to about 30%.
  • When the composition is a liquid or gel, a first unit dose is applied and held in place under the tongue for a predetermined time, for example for at least about 30 seconds, or more particularly about 60 seconds or more. A second unit dose may then be applied and held in place for a similar amount of time. Surprisingly, this procedure noticeably increases the effect of the composition of the invention in the treatment of agitation or the signs of agitation.
  • In another embodiment, the sublingual composition of Dexmedetomidine or a pharmaceutically acceptable salt thereof is a hard tablet or a compressed powder tablet. The tablet may conveniently be designed to dissolve under the tongue in about 30 to 120 seconds as disclosed in U.S. Pat. No. 6,221,392 to Khankari, et al., incorporated herein by reference. In a particular embodiment, the sublingual composition of Dexmedetomidine or a pharmaceutically acceptable salt thereof is a hard tablet having a low grit component for an organoleptically pleasant mouth feel. The tablet (or particles thereof containing the active ingredient which can be compressed to form the tablet) may also comprise a protective outer coating, e.g. any polymer conventionally used in the formation of microparticles, matrix-type microparticles and microcapsules.
  • In a further embodiment, the sublingual composition of Dexmedetomidine or a pharmaceutically acceptable salt thereof is a hard, compressed, rapidly dissolvable tablet. The tablet conveniently includes the active ingredient within a matrix. The matrix may be composed of, for example, at least one filler and a lubricant. Fillers include, for example, lactose or mannitol, and suitable lubricants include magnesium stearate, silicon dioxide and talc. The matrix may also include one or more of: a binder (e.g. povidone, a sugar or carboxymethylcellulose), a disintegrant (e.g. croscarmellose sodium, crospovidone or sodium starch glycolate), a sweeting agent (e.g. sucralose) and the like. The tablet may conveniently have a friability of about 2% or less and a hardness of about 15 to about 50 Newtons.
  • Another aspect of the present invention provides a method of making a packaged, sublingual tablet. The method includes the steps of: (a) forming a mixture comprising Dexmedetomidine or a pharmaceutically acceptable salt thereof and a matrix including at least a non-direct compression filler and a lubricant; (b) compressing the mixture to form a plurality of hard, compressed, rapidly disintegrable particles (e.g. beads) including the active ingredient distributed in the sublingually dissolvable matrix; and (c) storing the product in bulk prior to packaging. In another embodiment, the dosage forms are then packaged in a lumen of a package such that there are more than one per package. Direct compression is the preferred method of forming the dosage forms. There is also provided hereby an openable and reclosable package containing a plurality of hard, compressed, rapidly dissolving tablets adapted for direct oral dosing as described above.
  • In another embodiment, the present invention is a sublingual tablet comprising an effervescent agent. The effervescent agent may conveniently be present in an amount up to about 95% by weight, based on the weight of the finished tablet, and more particularly in an amount of between about 30% and about 80% by weight. Sufficient effervescent material is included in the tablet composition to generate more than about 5 cm3 but less that about 30 cm3 of gas upon exposure of the tablet to an aqueous environment. Sublingual compositions comprising effervescent agents are disclosed in U.S. Pat. No. 6,200,604, which is incorporated herein by reference.
  • In one particular embodiment, an effervescent agent releases carbon dioxide e.g., as a result of the reaction of a soluble acid source with an alkaline carbonate or bicarbonate. The acid source may conveniently include food acids and acids such as citric acid, tartaric, amalic, fumeric, adipic and succinic acid. Carbonate and bicarbonate sources include dry solid carbonate and bicarbonate salts such as sodium bicarbonate, sodium carbonate, potassium bicarbonate, potassium carbonate, magnesium carbonate and the like.
  • Spray compositions of the present invention for sublingual administration may include one or more pharmaceutically acceptable liquids (e.g., present in the amount of about 30% to about 99.99% by weight of the composition). Such liquids may be solvents, co-solvents, or non-solvents for Dexmedetomidine or a pharmaceutically acceptable salt thereof. Examples of pharmaceutically acceptable liquids include water, ethanol, dimethyl sulfoxide, propylene glycol, polyethylene glycol, propylene carbonate, pharmaceutically acceptable oils (e.g., soybean, sunflower, peanut, peppermint etc.) and the like. The pharmaceutically acceptable liquid is selected either to dissolve the active pharmaceutical ingredient, to produce a stable, homogenous suspension or solution of it, or to form any combination of a suspension or solution.
  • Furthermore, sublingual, spray formulations of Dexmedetomidine or a pharmaceutically acceptable salt thereof may include one or more carriers and/or excipients. Examples of carriers/excipients include viscosity-modulating materials (e.g. polymers, sugars, sugar alcohols, gums, clays, silicas, and the like). One particular polymer that may conveniently be used is polyvinylpyrrolidone (PVP). The viscosity-modulating material may conveniently be present in the amount of from about 0.01% to about 65% by weight of the spray formulation. Other examples of carriers/excipients include preservatives (e.g. ethanol, benzyl alcohol, propylparaben and methylparaben). Preservatives may conveniently be present in the amount of from about 0.001% to about 10% by weight of the spray formulation. Carriers/excipients may also be flavoring agents, sweeteners (e.g. sugars such as sucrose, glucose, dextrose, maltose, fructose, etc.), artificial sweeteners (e.g. saccharin, aspartame, acesulfame, sucralose etc.), or sugar alcohols (e.g. mannitol, xylitol, lactitol, maltitol syrup etc.) present conveniently in an amount of from about 0.001% to about 65% by weight of the spray formulation. Other examples of carriers/excipients include buffers and pH-adjusting agent (e.g., sodium hydroxide, citrate, and citric acid) conveniently present in an amount of from about 0.01% to about 5% by weight of the spray formulation. Coloring agents (e.g. present in an amount of from about 0.001% to about 5% by weight of the spray formulation), fragrances (e.g. present in an amount of from about 0.001% to about 1% by weight of the spray formulation), chelating agents such as EDTA (e.g. present in an amount of from about 0.001% to about 1% by weight of the spray formulation), UV absorbers (e.g. present in an amount of from about 0.001% to about 10% by weight of the spray formulation), and anti-foam agents (e.g. low molecular weight alcohols, dimethicone) conveniently present in an amount of from about 0.001% to about 5% by weight of the spray formulation may also be included as appropriate carriers/excipients in the spray formulations of the present invention.
  • One particular aspect of the present invention provides a sublingual film comprising Dexmedetomidine or a pharmaceutically acceptable salt thereof, together with one or more carriers and/or excipients, for the treatment of agitation.
  • Excipients which may be incorporated into the sublingual films of the present invention include one or more of the following: film forming agents, mouth feel improvers, plasticizers, stabilizers, surfactants, preservatives, sweetening agents, colorants, flavourants, emulsifiers, disintegrants, salivating agents, antioxidants, permeation enhancers, solvents and the like.
  • Film forming agents generally mean agents that provide structure to the film of the present invention. The effective amount of the film forming agent ranges from about 10% to about 99%, more preferably about 50% to about 90% by weight of the composition. Film forming agents that can be utilized as part of the film composition of the present invention include, but are not limited to, cellulose ethers, modified starches, natural gums, edible polymers, seaweed extracts, land plant extracts, pullulan, polyvinylpyrrolidone, derivatives thereof and combinations thereof.
  • Examples of cellulose ethers include, but are not limited to, methylhydroxycellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, carboxymethylcellulose, derivatives thereof and combinations thereof.
  • Modified starches include, but are not limited to, acid and enzyme hydrolyzed corn and potato starches, derivatives thereof and combinations thereof.
  • Examples of natural gums include, but are not limited to, gum arabic, guar gum, locust bean gum, carrageenan gum, acacia, karaya, ghatti, tragacanth agar, tamarind gum, xanthan gum, derivatives thereof and combinations thereof.
  • Examples of edible polymers include, but are not limited to, microcrystalline cellulose, cellulose ethers, xanthan, derivatives thereof and combinations thereof.
  • Seaweed extract examples include, but are not limited to, sodium alginate, carrageenans, derivatives thereof and combinations thereof.
  • Land plant extracts include, but are not limited to, konjac, pectin, arabinogalactan, derivatives thereof and combinations thereof.
  • Particular film forming agents include pullulan, sodium alginate, polyvinylpyrrolidone, methylcellulose and methylhydroxycellulose (MHC).
  • The term “solvent” generally refers to liquids that will dissolve solutes. A solvent may be used to dissolve film-forming agents and other excipients to prepare film-forming compositions of the present invention. Solvents include, but are not limited to, demineralized/distilled water, ethyl alcohol, isopropyl alcohol, methyl ethyl ketone, propylene glycol methyl ether acetate, dimethyl acetamide, ethylene glycol mono-propyl ether, and toluene. A sublingual film of the present invention may conveniently comprise a solvent in an amount up to about 1% w/w.
  • The term “stabilizer” generally refers to an agent that will impart stability to the formulation during its shelf life. Stabilizers of the present invention can include, for example, oil/water emulsifiers and flavor fixatives. The effective amount of a stabilizer agent in a composition of the invention may be, for example, in the range of about 0% to about 45%, more particularly about 4% to about 25%, by weight of the composition. Examples of suitable stabilizing agents of the present invention include, but are not limited to, gum arabic, microcrystalline cellulose, carrageenan, xanthan gum, locust bean gum, derivatives thereof and combinations thereof. Particular stabilizing agents of the present invention include gum arabic and microcrystalline cellulose.
  • “Disintegrants” can aid the dissolution of edible films allowing for the efficacy of the film to be realized sooner. Suitable disintegrants for use in an edible film of the present invention include, but are not limited to, alginic acid, microcrystalline cellulose and carboxymethylcellulose. Special disintegrants known as super-disintegrants are also suitable for use in an edible film of the present invention. Super-disintegrants include cross-linked polymers (e.g. crospovidone), cross-linked starches (e.g. sodium starch glycolate), and cross-linked celluloses (e.g. a modified carboxymethylcellulose such as croscarmellose). These super-disintegrants are insoluble in water and most other solvents, have rapid swelling properties, and have good water uptake with high capillary action, resulting in fast disintegration. Their insolubility in many solvents also means they enable the manufacture of sublingual compositions of this invention in a single step process as opposed to costly multi-step processes.
  • The disintegrants or super-disintegrants are conveniently present in a sublingual composition of this invention (e.g. an edible film) in an amount ranging from about 1% to about 10%, more particularly about 1% to about 5% by weight of the composition.
  • “Emulsifiers” suitable for use in an edible film of the present invention include, but are not limited to, gum arabic, carrageenan, triethanolamine stearate, quaternary ammonium compounds, acacia, gelatin, lecithin, bentonite, veegum, derivatives thereof and combinations thereof. Emulsifiers can be used in a composition of the present invention in an amount up to about 40%, more particularly up to about 25%, by weight of the composition. The emulsifier can be a stabilizer creating an oil/water emulsion encapsulating volatile oils and flavoring agents, thereby essentially acting as a flavor fixative. A particular emulsifier for use in an edible film of the present invention is gum arabic.
  • A “plasticizing agent” or “plasticizer” may be utilized to improve flexibility and reduce brittleness of an edible film composition of the present invention. The plasticizing agent may conveniently constitute up to about 30%, e.g. up to about 15% by weight of the composition. Examples of suitable plasticizing agents include, but are not limited to, glycerin, sorbitol, triacetin, monoacetin, diacetin, polyethylene glycol, propylene glycol, hydrogenated starch hydrolysates, corn syrups, low molecular weight propylene glycols, phthalate derivatives like dimethyl, diethyl and dibutyl phthalate, citrate derivatives such as tributyl, triethyl, acetyl citrate and castor oil derivatives thereof and combinations thereof. Particular plasticizing agents of the present invention include sorbitol and glycerin.
  • The term “preservative” generally refers to an excipient used to kill microorganisms or prevents, inhibits or retards their growth and reproduction, and is included in a product in a concentration only sufficient to prevent spoilage or the growth of inadvertently added microorganisms. Suitable preservative includes, but are not limited to, methylparaben, propylparaben and sodium benzoate. The preservative may conveniently be present in the composition from about 0.001% to about 10% w/w of the composition.
  • The term “sweetening agent” generally refers to an excipient used to impart sweetness to a pharmaceutical composition. Suitable sweetening agents for use in a composition of the present invention include, but are not limited to, aspartame, dextrose, glycerin, mannitol, saccharin sodium, sorbitol and sucrose. The sweetening agent may conveniently be present in the composition in an amount of from about 5% to about 20% w/w of the composition.
  • The term “coloring agent” or “colorant” generally refers to an excipient used to impart color to a pharmaceutical composition. Suitable colorants include, but are not limited to, FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, other F.D. & C. dyes, caramel, red ferric oxide, and natural coloring agents such as grape skin extract, beet red powder, beta-carotene, annatto, carmine, turmeric or paprika. The colorant may conveniently be present in the composition in an amount of from about 0.001% to about 10% w/w of the composition.
  • The term “flavoring agent” or “flavorant” generally refers to an excipient used to impart a pleasant flavor (and often also odor) to a pharmaceutical composition. Suitable flavorants include, but are not limited to, synthetic flavoring oils, flavoring aromatics, natural oils, extracts from whole plants or parts thereof such as leaves, flowers, fruits or combinations thereof. Examples include cinnamon oil, wintergreen oil, peppermint oil, clove oil, bay oil, anise oil, eucalyptus oil, thyme oil, cedar leave oil, nutmeg oil, sage oil, bitter almond oil and cassia oil. Other useful flavorants include vanilla, citrus fruit oils such as lemon, orange, grape, lime or grapefruit oil, and fruit essences such as apple, pear, peach, strawberry, raspberry, cherry, plum, pineapple or apricot essence. Flavorants of particular interest for use in a composition of the present invention include commercially available orange, grape, cherry and bubble gum flavors and mixtures thereof. The amount of flavoring used will depend on a number of factors, including the organoleptic effect desired. Particular flavorants include grape and cherry flavors, and citrus fruit flavors such as orange flavor. The flavorant may conveniently be present in the composition in an amount of from about 0.001% to about 10% w/w of the composition.
  • The term “salivating agent” is an agent that promotes greater salivation during use of a composition of the present invention. This may be an important feature if the composition is intended to be taken by the patient without the aid of water to help in the transporting of the composition to the stomach of the patient. The salivating agent can be, for example, an emulsifier or a food acid that initiates salivation in the mouth of the patient. Examples of emulsifiers useful as salivating agents include alkyl aryl sulfonates, alkyl sulfates, sulfonated amides and amines, sulfated and sulfonated esters and ethers, alkyl sulfonates, polyethoxylated esters, mono-, di-, and triglycerides, diacetyl tartaric esters of monoglycerides, polyglycerol esters, sorbitan esters and ethoxylates, lactylated esters, phospholipids such as lecithin, polyoxyethylene sorbitan esters, propylene glycol esters, sucrose esters, and mixtures thereof. The emulsifier may be either saturated or unsaturated. It should be noted that some of the emulsifiers that are salivating agents may also function as binders. Examples of food acids useful as salivating agents include citric acid, malic acid, tartarate, food salts such as sodium chloride and salt substitutes, potassium chloride, and mixtures thereof. The amount of salivating agent present in a sublingual film of the present invention may convenient be up to about 15% by weight of the final composition, e.g. in the range of from about 0.3% to 0.4% by weight of the composition.
  • The term “antioxidant” generally refers to an excipient used to inhibit oxidation and thus prevent deterioration of active agents by oxidative processes. Suitable antioxidants include, for example, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothio-glycerol, propyl gallate, sodium ascorbate, citric acid, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, EDTA and sodium edetate. The anti-oxidant may conveniently be present in the composition in an amount of from about 0.001% to about 2% w/w of the composition.
  • The term “permeation enhancer” generally refers to an excipient used to enhance permeation of an active agent to cellular membranes or enhance the local/systemic absorption of the active agent. Permeation enhancers that may be used in the present invention include, but are not limited to, solubilizers such as alcohols, polyethylene glycols, chelating agents (e.g. cyclodextrins), sucrose laurate or sucrose oleate. The permeation enhancer may conveniently be present in the composition in an amount of from about 0.1% to about 5% w/w of the composition.
  • In one embodiment of the present invention, the sublingual pharmaceutical composition of the present invention includes a mucosal permeation enhancer appropriate for enhancing the mucosal absorption of the composition.
  • Sublingual Dexmedetomidine formulations (such as sprays, drops, and the like) may be made by mixing appropriate quantities of the foregoing ingredients in accordance with standard good manufacturing practices. The relative amounts of each ingredient should not interfere with the desirable pharmacological and pharmacokinetic properties of the resulting formulation.
  • Sublingual Dexmedetomidine films of the present invention may be conveniently prepared using PharmFilm® technology (owned by MonoSol) or technology owned by ARx LLC. Various patents and patent applications are incorporated herein in entirety and includes U.S. Pat. Nos. 9,585,961, 7,470,397, 7,727,466, 9,248,146, 9,545,376, 2017-0087084, 9,662,297, 9,662,301, 2017-0246108, 2017-0252294, 9,441,142 assigned to ARx LLC and U.S. Pat. Nos. 7,425,292, 7,357,891, 8,663,687, 8,685,437, 7,897,080, 8,241,661, 8,617,589, 8,936,825, 9,561,191, 9,303,918, 9,346,601, 8,282,954, 7,972,618, 9,073,294 assigned to Monosol Rx.
  • In preparing the sublingual film of the present invention the active agent, e.g. Dexmedetomidine or a pharmaceutically acceptable salt thereof, film forming agents and optionally one or more carriers and/or excipients selected from the group comprising of mouth feel improver, plasticizer, stabilizer, surfactant, preservative, sweetening agent, colorant, flavourant, emulsifier, disintegrant, salivating agent, antioxidant, permeation enhancer are dissolved in a compatible solvent to form a film forming composition. Compatible solvents include water, alcohols such as ethanol, ethyl acetate, acetone, and mixtures thereof. The film forming composition is cast on a releasable carrier and dried to form a sheet/film. The carrier material must have a surface tension which allows the film solution to spread evenly across the intended carrier width without soaking to form a destructive bond between the film carrier substrates. Examples of suitable carrier materials include glass, stainless steel, Teflon and polyethylene-impregnated paper. Drying of the film may be carried out at high temperature using a drying oven, drying terminal, vacuum drier, or any other suitable drying equipment which does not adversely affect the ingredients of which the film is composed. The sublingual film of the present invention can also be prepared by other established processes e.g. extrusion (for example, Hot melt extrusion, Solid dispersion extrusion), casting (for example, solid casting or semi-solid casting), Rolling methods and the like.
  • V. Administration
  • In an aspect, the present invention provides a sublingual composition comprising an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof, administered to a subject in an amount sufficient to effectively treat agitation. The amount of alpha-2 adrenergic agonist is sufficient to effectively treat agitation without causing significant sedation. The alpha-2 adrenergic agonist may conveniently be delivered on an “as needed basis” in one, two or more doses per day to the animal (e.g. human) subject. The composition may also be administered via a single dosage form or via multiple dosage forms.
  • In another aspect, the present invention provides a sublingual composition comprising Dexmedetomidine or a pharmaceutically acceptable salt thereof, administered to a subject in an amount sufficient to effectively treat agitation. In a particular aspect, the amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof used is sufficient to effectively treat agitation without causing significant sedation. The Dexmedetomidine or a pharmaceutically acceptable salt thereof may conveniently be delivered on an “as needed basis” in one, two or more doses per day to the animal (e.g. human) subject. The composition may also be administered via a single dosage form or via multiple dosage forms.
  • Following administration of a composition of this invention to a subject, a therapeutic (i.e. anti-agitation) effect may begin within about 60 minutes (e.g. within about 30, 20, 15, 10, 5, 3, 2 or 1 minutes) after administration, or within about 30 seconds after administration. The signs of agitation may also be relieved within about 1 to about 60 minutes after administration, and more typically within about 5 to about 30 minutes. A second dose of the composition of this invention may be administered to the subject if the signs of agitation are not relieved within about 60 minutes.
  • Treatment protocols may include one or more dosage intervals (e.g. two or more dosage intervals, five or more dosage intervals, or ten or more dosage intervals). Depending on the physiology of the subject and the desired therapeutic effect, the duration of dosage intervals and treatment protocols according to embodiments of the present invention may vary.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may be administered as a sublingual composition to treat agitation or the signs of agitation either alone or in combination with one or more further active agents. When used in combination, the active agents can either be formulated as a single composition or as two or more separate compositions, which can be administered simultaneously, sequentially or separated by an appropriate period of time.
  • Where Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered with a second active agent to treat agitation or the signs of agitation, the weight ratio of respectively Dexmedetomidine or a pharmaceutically acceptable salt thereof to the second active agent may generally be in the range from about 1:2 to about 1:2.5; about 1:2.5 to about 1:3; about 1:3 to about 1:3.5 about 1:3.5 to about 1:4; about 1:4 to about 1:4.5, about 1:4.5 to about 1:5; about 1:5 to about 1:10; and about 1:10 to about 1:25. For example, the weight ratio may particularly be between about 1:1 to about 1:5; about 1:5 to about 1:10; about 1:10 to about 1:15; or about 1:15 to about 1:25. Alternatively, the weight ratio of respectively the second active agent to Dexmedetomidine or a pharmaceutically acceptable salt may be in the range of from about 2:1 to about 2.5:1; about 2.5:1 to about 3:1; about 3:1 to about 3.5:1; about 3.5:1 to about 4:1; about 4:1 to about 4.5:1; about 4.5:1 to about 5:1; about 5:1 to about 10:1; and about 10:1 to about 25:1. For example, the weight ratio of respectively the second active agent to Dexmedetomidine or a pharmaceutically acceptable salt thereof may particularly be in the range of from about 1:1 to about 5:1; about 5:1 to about 10:1; about 10:1 to about 15:1; or about 15:1 to about 25:1. It is to be understood that all ranges between the quoted ranges are also covered herein, and constitute further particular aspects of this invention.
  • VI. Dosing Regimen
  • The dosing regimen employed may depend on several factors, such as the type of agitation treated, the severity of the signs, and whether the agitation is due to an underlying medical condition.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may be administered sublingually in any appropriate dose to an animal (e.g. human). In certain embodiments, the human dose may be from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms). The dose may be administered one or more times a day.
  • Dexmedetomidine or a pharmaceutically acceptable salt thereof may be administered sublingually in any appropriate dose to a human. In some variations, the human dose may be from about 0.05 micrograms/kg weight of subject to about 1.5 micrograms/kg weight of subject. Examples of suitable dosages include: about 0.1 micrograms/kg to about 1 micrograms/kg, about 0.1 micrograms/kg to about 0.5 micrograms/kg, about 0.1 micrograms/kg to about 0.4 micrograms/kg, about 0.1 micrograms/kg to about 0.3 micrograms/kg, about 0.1 micrograms/kg to about 0.2 micrograms/kg, about 0.07 micrograms/kg, about 0.05 micrograms/kg, about 0.1 micrograms/kg, about 0.2 micrograms/kg, about 0.3 micrograms/kg, about 0.4 micrograms/kg, about 0.5 micrograms/kg, about 0.6 micrograms/kg, about 0.7 micrograms/kg, about 0.8 micrograms/kg, about 0.9 micrograms/kg, about 1.0 micrograms/kg, about 1.1 micrograms/kg, about 1.2 micrograms/kg, about 1.3 micrograms/kg, about 1.4 micrograms/kg, about 1.5 micrograms/kg. The dose may be administered one or more times a day.
  • VII. Specific Embodiments of the Invention
  • Embodiment 1. A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering sublingually to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • Embodiment 2. A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, wherein said Dexmedetomidine or a pharmaceutically acceptable salt thereof is sublingually administered at a dosage that treats agitation or the signs of agitation without causing significant sedation.
  • Embodiment 3. The method according to Embodiment 1 or 2, wherein said dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof is in range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 4. The method according to Embodiments 1, 2 or 3, wherein said subject is mammal, preferably human.
  • Embodiment 5. The method according to any one of Embodiments 1 to 4, wherein said agitation is associated with a neurodegenerative disease.
  • Embodiment 6. The method according to Embodiment 5, wherein said neurodegenerative disease is selected from Alzheimer disease, fronto-temporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, and progressive supranuclear palsy.
  • Embodiment 7. The method according to any one of Embodiments 1 to 4, wherein the agitation is associated with a neuropsychiatric condition.
  • Embodiment 8. The method according to Embodiment 7, wherein said neuropsychiatric condition is selected from schizophrenia, bipolar illness (such as mania, disorder), delirium, and depression.
  • Embodiment 9. The method according to Embodiment 5, wherein the agitation is associated with sundown syndrome in dementia or Alzheimer's disease.
  • Embodiment 10. A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is not perioperative agitation and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 11. A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with a neurodegenerative disease and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 12. A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with a neuropsychiatric condition and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 13. A sublingual composition for use in the treatment of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with sundown syndrome in dementia or Alzheimer's disease and said sublingual composition comprises an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers/excipients.
  • Embodiment 14. The sublingual composition according to Embodiment 11, wherein said neurodegenerative disease is selected from the group consisting of Alzheimer disease, frontotemporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis Creutzfeldt-Jakob disease, multiple system atrophy, and progressive supranuclear palsy.
  • Embodiment 15. The sublingual composition according to Embodiment 14, wherein said neurodegenerative disease is selected from dementia, frontotemporal dementia, Alzheimer's disease and Parkinson's disease.
  • Embodiment 16. The sublingual composition according to Embodiment 12, wherein said neuropsychiatric condition is selected from the group consisting of schizophrenia, bipolar illness (such as mania, disorder), delirium and depression.
  • Embodiment 17. The sublingual composition according to any one of Embodiments 10 to 16, wherein said composition is selected from a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • Embodiment 18. The sublingual composition according to Embodiment 17, wherein the composition is a film.
  • Embodiment 19. The sublingual composition according to Embodiment 18, wherein the film is mucoadhesive in nature and provides a quick onset of action.
  • Embodiment 20. The sublingual composition according to any one of Embodiments 10 to 19, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered at a dosage that treats agitation or the signs of agitation without causing significant sedation.
  • Embodiment 21. The sublingual composition according to Embodiment 20 wherein the observed level of sedation is not greater than 3 on the Ramsay sedation scale.
  • Embodiment 22. The sublingual composition according to any one of Embodiments 10 to 21, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered to said subject (e.g. human) at a dosage in the range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 23. A method of treating agitation or the signs of agitation in a subject in need thereof, the method comprises sublingually administering to said subject an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof, wherein the agitation is not perioperative agitation.
  • Embodiment 24. The method according to Embodiment 23, wherein the agitation is associated with a neurodegenerative disease and/or neuropsychiatric condition.
  • Embodiment 25. The method according to Embodiment 24, wherein the neurodegenerative disease is selected from the group consisting of Alzheimer disease, frontotemporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, progressive supranuclear palsy or other related neurodegenerative disorder.
  • Embodiment 26. The method according to Embodiment 24, wherein the neuropsychiatric condition is selected from the group consisting of schizophrenia, bipolar illness (e.g. bipolar disorder or bipolar mania), delirium and depression.
  • Embodiment 27. The method according to any one of Embodiments 23 to 26, wherein agitation or the signs of agitation are treated effectively without causing significant sedation.
  • Embodiment 28. The method according to any one of Embodiments 23 to 27, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered in form of a film, wafer, patch, lozenge, gel, spray, tablet, liquid drops or the like.
  • Embodiment 29. A method of treating of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures), and said method comprises administering to said subject sublingually an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • Embodiment 30. A method of treating of agitation or the signs of agitation in a subject in need thereof, wherein said agitation is associated with an alcohol and substance abuse withdrawal and said method comprises administering to said subject sublingually an effective amount of Dexmedetomidine or a pharmaceutically acceptable salt thereof.
  • Embodiment 31. The method according to Embodiment 29 or 30, wherein said Dexmedetomidine or a pharmaceutically acceptable salt thereof is sublingually administered at a dosage that treats said agitation or the signs of agitation without causing significant sedation.
  • Embodiment 32. The method according to Embodiment 31, wherein said dosage of Dexmedetomidine or a pharmaceutically acceptable salt thereof is in range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 33. The composition or method according to any preceding Embodiment, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered once, twice or thrice daily or on an “as needed” basis.
  • Embodiment 34. The composition or method according to any preceding Embodiment, wherein Dexmedetomidine or a pharmaceutically acceptable salt thereof is administered in a manner that produces a therapeutic effect in less than about 60 minutes, particularly within about 30 seconds to about 30 minutes.
  • Embodiment 35. A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering sublingually to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof.
  • Embodiment 36. A method of treating agitation or the signs of agitation in a subject in need thereof comprising administering to said subject an effective amount of an alpha-2 adrenergic agonist or a pharmaceutically acceptable salt thereof, wherein said alpha-2 adrenergic agonist is sublingually administered at a dosage that treats agitation or the signs of agitation without causing significant sedation.
  • Embodiment 37. The method according to Embodiment 35 or 36, wherein said dosage of alpha-2 adrenergic agonist is in range of from about 3 micrograms to about 100 micrograms (e.g. about 5 micrograms to about 100 micrograms, about 5 micrograms to about 90 micrograms, about 5 micrograms to about 85 micrograms, about 5 micrograms to about 80 micrograms, about 5 micrograms to about 75 micrograms, about 5 micrograms to about 70 micrograms, about 5 micrograms to about 65 micrograms, about 5 micrograms to about 60 micrograms, about 5 micrograms to about 55 micrograms, about 5 micrograms to about 50 micrograms, about 5 micrograms to about 45 micrograms, about 5 micrograms to about 40 micrograms, about 5 micrograms to about 35 micrograms, about 5 micrograms to about 30 micrograms, about 5 micrograms to about 25 micrograms, about 5 micrograms to about 20 micrograms, about 5 micrograms to about 15 micrograms, about 5 micrograms to about 10 micrograms, less than 10 micrograms (e.g. about 5, 6, 7, 8, or 9 micrograms), about 10 micrograms, about 12 micrograms, about 14 micrograms, about 15 micrograms, about 16 micrograms, about 18 micrograms, about 20 micrograms, about 30 micrograms, about 50 micrograms).
  • Embodiment 38. The method according to Embodiments 35, 36 or 37, wherein said subject is mammal, preferably human.
  • Embodiment 39. The method according to any one of Embodiments 35 to 38, wherein said agitation is associated with a neurodegenerative disease.
  • Embodiment 40. The method according to Embodiment 39, wherein said neurodegenerative disease is selected from Alzheimer disease, fronto-temporal dementia (FTD), dementia, dementia with Lewy bodies (DLB), post-traumatic stress disorder, Parkinson's disease, vascular dementia, vascular cognitive impairment, Huntington's disease, multiple sclerosis, Creutzfeldt-Jakob disease, multiple system atrophy, and progressive supranuclear palsy.
  • Embodiment 41. The method according to any one of Embodiments 35 to 38, wherein the agitation is associated with a neuropsychiatric condition.
  • Embodiment 42. The method according to Embodiment 41, wherein said neuropsychiatric condition is selected from schizophrenia, bipolar illness (such as mania, disorder), delirium, and depression.
  • Embodiment 43. The method according to Embodiment 39, wherein the agitation is associated with sundown syndrome in dementia or Alzheimer's disease.
  • Embodiment 44. The method according to any one of Embodiments 35 to 38, wherein said agitation is associated with an OPD/IPD procedure (e.g. MRI, CT or CAT scan, lumbar puncture, bone marrow aspiration/biopsy, tooth extraction or other dental procedures).
  • Embodiment 45. The method according to any one of Embodiments 35 to 38, wherein said agitation is associated with an alcohol and substance abuse withdrawal.
  • Embodiment 46. The method according to any one of Embodiments 35 to 38, wherein said alpha-2 adrenergic agonist include, but is not limited to, Clonidine, Guanfacine, Guanabenz, Guanoxabenz, Guanethidine, Xylazine, Tizanidine, Medetomidine, Dexmedetomidine, Methyldopa, Methylnorepinephrine, Fadolmidine Iodoclonidine, Apraclonidine, Detomidine, Lofexidine, Amitraz, Mivazerol, Azepexol, Talipexol, Rilmenidine, Naphazoline, Oxymetazoline, Xylometazoline, Tetrahydrozoline, Tramazoline, Talipexole, Romifidine, propylhexedrine, Norfenefrine, Octopamine, Moxonidine, Lidamidine, Tolonidine, UK14304, DJ-7141, ST-91, RWJ-52353, TCG-1000, 4-(3-aminomethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione, and 4-(3-hydroxymethyl-cyclohex-3-enylmethyl)-1,3-dihydro-imidazole-2-thione or a pharmaceutically acceptable salt thereof.
  • Embodiment 47. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein said agitation is associated with neurodegenerative diseases and said method comprises administering to said subject a therapeutically effective amount of Dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 48. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein said agitation is associated with sundowning syndrome in dementia or Alzheimer's disease and said method comprises: administering to said subject a therapeutically effective amount of Dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 49. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, intoxication and other neuropsychiatric conditions and said method comprises: administering to said subject a therapeutically effective amount of Dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 50. The method as embodied in embodiment 47, wherein said neurodegenerative disease is Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile x syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata.
  • Embodiment 51. The method as embodied in embodiment 50, wherein said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • Embodiment 52. The method as embodied in embodiments 47 to 49, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by oromucosal route including sublingual or buccal route in the form of pharmaceutical composition.
  • Embodiment 53. The method as embodied in embodiment 52, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by sublingual route.
  • Embodiment 54. The method as embodied in embodiments 47 to 49, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject (i.e. Human) is in a range of about 1 μg to about 105 μg, preferably 3 μg to 100 μg, more preferably 5 μg to 50 μg.
  • Embodiment 55. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with neurodegenerative diseases and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 56. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, post-traumatic stress disorder, alcohol and substance abuse withdrawal, intoxication and other neuropsychiatric conditions and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 57. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with sundowning syndrome in dementia or Alzheimers disease and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 58. A pharmaceutical composition for use in the treatment, prevention or reduction in the symptoms of agitation in a subject in need thereof wherein said chronic agitation is associated with neurodegenerative diseases and said pharmaceutical composition comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 59. A pharmaceutical composition for use in the treatment, prevention or reduction in the symptoms of agitation in a subject in need thereof wherein said agitation is associated with schizophrenia, bipolar disorder, bipolar mania, delirium, intoxication and other neuropsychiatric conditions and said pharmaceutical composition comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 60. A pharmaceutical composition for use in the treatment, prevention or reduction in the symptoms of agitation in a subject in need thereof wherein said agitation is associated with sundowning syndrome in dementia or Alzheimer's disease and said pharmaceutical composition comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 61. The method/composition as embodied in embodiments 55 and 58, wherein said neurodegenerative disease is selected from Frontotemporal dementia, Dementia, Alzheimer's disease, Parkinson's disease Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile x syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morpervan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata.
  • Embodiment 62. The method/composition as embodied in embodiment 61, wherein said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • Embodiment 63. The method/composition as embodied in embodiments 55 to 60, wherein said pharmaceutical composition is administered to said subject by an oromucosal route including sublingual or buccal route in the form of pharmaceutical composition.
  • Embodiment 64. The method/composition as embodied in embodiment 63, wherein said pharmaceutical composition is a tablet, film, gel etc.
  • Embodiment 65. The method/composition as embodied in embodiments 55 to 60, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject (i.e. Human) is in a range of about 1 μg to about 105 μg, preferably about 3 μg to 100 μg, more preferably about 5 μg to 50 μg and most preferably less than 10 μg.
  • Embodiment 66. Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof composition for treating a subject suffering from agitation associated with neurodegenerative disease.
  • Embodiment 67. Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof composition for treating a subject suffering from agitation associated with neuropsychiatric conditions including schizophrenia, bipolar disorder, bipolar mania, delirium, intoxication and other neuropsychiatric conditions.
  • Embodiment 68. Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof composition for treating a subject suffering from agitation associated with sundowning syndrome in dementia or Alzheimer's disease.
  • Embodiment 69. The use as embodied in embodiment 66, wherein neurodegenerative disease is selected from Frontotemporal dementia, dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile x syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata.
  • Embodiment 70. The use as embodied in embodiment 69, wherein said neurodegenerative disease is preferably Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease.
  • Embodiment 71. A sublingual film composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with neurodegenerative disease.
  • Embodiment 72. A sublingual film composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with neuropsychiatric conditions such as bipolar disorder, schizophrenia. bipolar mania. Delirium, intoxication and other neuropsychiatric conditions.
  • Embodiment 73. A sublingual film composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with sundowning syndrome in dementia or Alzheimer's disease.
  • Embodiment 74. A method of treating, preventing or reducing the symptoms of aggression in a subject in need thereof and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 75. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, the method comprises administering to said subject dexmedetomidine, or a pharmaceutically acceptable salt thereof through sublingual film in a dose range of 1 μg to 105 μg, preferably about 3 μg to 100 μg and more preferably about 5 μg to 50 μg, more preferably about 5 μg to 20 μg, most preferably less than 10 μg.
  • Embodiment 76. The method as embodied in embodiment 75, wherein the agitation is acute agitation.
  • Embodiment 77. The method as embodied in embodiment 75, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 78. The method as embodied in embodiment 77, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-Lemli Opitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability and Agrypnia excitata, sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Frontotemporal dementia, Alzheimer's disease or Parkinson's disease.
  • Embodiment 79. The method as embodied in embodiment 77, wherein the neuropsychiatric conditions is selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal or intoxication.
  • Embodiment 80. The method embodied in embodiment 75, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to the said subject in a dose ranging from about 1 μg to about 105 μg; preferably from about 3 μg to about 100 μg, more preferably about 5 μg to about 50 μg, most preferably about 5 μg to about 20 μg.
  • Embodiment 81. A sublingual film comprising about 5 μg to about 100 μg of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation in a subject.
  • Embodiment 82. The sublingual film as embodied in embodiment 81, wherein the agitation is acute agitation.
  • Embodiment 83. The sublingual film as embodied in embodiment 82, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 84. The sublingual film as embodied in embodiment 83, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-LemliOpitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability, Agrypnia excitata, sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Frontotemporal dementia, Alzheimer's disease or Parkinson's disease.
  • Embodiment 85. The sublingual film as embodied in embodiment 83, wherein the neuropsychiatric conditions is selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal or intoxication.
  • Embodiment 86. The sublingual film as embodied in embodiment 81, wherein the film comprises dexmedetomidine, or a pharmaceutically acceptable salt thereof ranging from about 1 μg to about 105 μg; preferably from about 3 μg to about 100 μg, more preferably about 5 μg to about 50 μg and most preferably about 5 μg to about 20 μg and less than 10 μg.
  • Embodiment 87. Use of dexmedetomidine, or a pharmaceutically acceptable salt thereof for treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein the agitation or aggression is not perioperative agitation.
  • Embodiment 88. The use as embodied in embodiment 87, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 89. The use as embodied in embodiment 88, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-LemliOpitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability, Agrypnia excitata, sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Frontotemporal dementia, Alzheimer's disease and Parkinson's disease.
  • Embodiment 90. The use as embodied in embodiment 88, wherein the neuropsychiatric conditions are selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal and intoxication.
  • Embodiment 91. The use as embodied in embodiment 87, wherein the agitation is acute agitation.
  • Embodiment 92. The use as embodied in embodiment 87, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by oromucosal route preferably by sublingual or buccal route.
  • Embodiment 93. The use as embodied in embodiment 92, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered in form of pharmaceutical composition selected from the group comprising of tablet, film and gel.
  • Embodiment 94. The use as embodied in embodiment 87, wherein Dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to the said subject in a dose ranging from about 1 μg to about 105 μg; preferably from about 3 μg to about 100 μg, more preferably about 5 μg to about 50 μg, most preferably about 5 μg to about 20 μg and most preferably less than 10 μg.
  • Embodiment 95. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof by sublingual route, wherein the agitation is not perioperative agitation.
  • Embodiment 96. A sublingual pharmaceutical composition for treating, preventing or reducing the symptoms of agitation in a subject in need thereof, the composition comprises a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and one or more pharmaceutical excipient, wherein the agitation is not perioperative agitation.
  • Embodiment 97. A sublingual use of dexmedetomidine, or a pharmaceutically acceptable salt thereof for treating, preventing or reducing the symptoms of agitation in a subject in need thereof, wherein the agitation is not perioperative agitation.
  • Embodiment 98. A sublingual composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with neurodegenerative disease.
  • Embodiment 99. A sublingual composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with bipolar disorder, schizophrenia. bipolar mania. Delirium, intoxication and other neuropsychiatric conditions.
  • Embodiment 100. A sublingual composition of dexmedetomidine, or a pharmaceutically acceptable salt thereof for use in the treatment of agitation associated with sundowning syndrome in dementia or Alzheimer's disease.
  • Embodiment 101. A method of treating, preventing or reducing the symptoms of agitation which is other than perioperative agitation in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof by sublingual route.
  • Embodiment 102. A sublingual film for treating, preventing or reducing the symptoms of agitation which is other than perioperative agitation in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof.
  • Embodiment 103. A method of treating, preventing or reducing the symptoms of aggression in a subject in need thereof and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 104. The method as embodied in embodiment 96, wherein the agitation is associated with an illness including neurodegenerative disease and/or neuropsychiatric conditions.
  • Embodiment 105. The method as embodied in embodiment 102, wherein the neurodegenerative disease is selected from the group consisting of neurodegenerative disease Dementia, Frontotemporal dementia, Alzheimer's disease, Parkinson's disease, Multiple system atrophy, Creutzfeldt-jakob disease, Corticobasal degeneration, Gerstmannstraussler-scheinker syndrome, Huntington disease, Fatal familial insomnia, Cushing's syndrome, Hypercortisolism, Neurofibromatosis type 1, Norrie disease, Progressive supranuclear palsy, Hereditary spastic paraplegia, Alpers syndrome, Smith-LemliOpitz syndrome, Fragile X syndrome, Mulvihill-smith syndrome, Transmissible spongiform encephalopathy, Morvan syndrome, Morvan's fibrillary chorea, Peripheral nerve hyperexcitability, Agrypnia excitata, sundowning syndrome in dementia or sundowning syndrome in Alzheimer's disease, preferably Dementia, Frontotemporal dementia, Alzheimer's disease or Parkinson's disease.
  • Embodiment 106. The method as embodied in embodiment 102, wherein the neuropsychiatric conditions is selected from the group consisting of schizophrenia, bipolar disorder, bipolar mania, delirium post-traumatic stress disorder, alcohol and substance abuse withdrawal or intoxication.
  • Embodiment 107. The method as embodied in embodiment 96, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered to said subject by oromucosal route preferably by sublingual or buccal route.
  • Embodiment 108. The method as embodied in embodiment 105, wherein dexmedetomidine, or a pharmaceutically acceptable salt thereof is administered in form of pharmaceutical composition selected from the group comprising of tablet, film or gel.
  • Embodiment 109. A method of treating, preventing or reducing the symptoms of agitation in a subject in need thereof wherein said agitation is associated with other conditions such as OPD/IPD Procedures like MRI, CT or CAT Scan, Lumbar Puncture, Bone marrow aspiration/biopsy, Tooth extraction or other dental procedures and said method comprises administering to said subject a pharmaceutical composition comprising a therapeutically effective amount of dexmedetomidine, or a pharmaceutically acceptable salt thereof and pharmaceutical acceptable excipient(s).
  • Embodiment 110. The methods/composition as embodied in the preceding embodiments, wherein the pharmaceutical composition includes intranasal formulations, parenteral formulations, oral formulations, transdermal formulations, liposomal formulations and the like.
  • The Embodiments hereinabove are not intended to be limiting, and, in practicing the present invention, alternative or additional Embodiments may be provided.
  • VII. Examples
  • The following Examples are intended to be illustrative and not limiting.
  • Example 1
  • Formulation 1: Sub-Lingual Tablet
  • TABLE 1
    Composition for a typical Sub-lingual tablet formulation
    used for sublingual delivery
    Ingredients Quantity Ranges
    Dexmedetomidine HCl 50
    (equivalent to base) micrograms
    Povidone 5.0 mg 1.0-10.0%
    Croscarmellose Sodium 7.0 mg    5-15%
    Sucralose 1.0 mg 0.05-3.0%
    Magnesium Stearate 0.75 mg   0.1-2.0%
    Talc 0.75 mg   0.1-2.0%
    Mannitol q.s 75.0 mg q.s. 100%
    Water q.s
  • Manufacturing Process
  • Dexmedetomidine hydrochloride and excipients such as binder and sweetener are dissolved/dispersed into a pharmaceutically acceptable solvent (preferably water) and this solution is used to granulate the sifted blend of all other ingredients except lubricant and glidant in suitable mixer/granulator. The granules are then dried in a fluid-bed drier or other suitable one such as tray drier. The dried granules are then sized appropriately in quadro-co-mill or multi-mill. The sized granules are then loaded into a suitable blender such as V-blender and lubricated with Magnesium stearate and Talc and then the final lubricated blend is then used for compressing into tablets of specific dimensions using appropriate tooling.
  • Formulation 2: Sub-Lingual Film
  • TABLE 2
    Composition for a typical Sub-lingual film formulation
    used for sublingual delivery
    Ingredients Quantity Ranges
    Dexmedetomidine HCl 50
    (equivalent to base) micrograms
    Polyethylene oxide 5.0 mg   3-25%
    Polyethylene Glycol 5.0 mg   3-25%
    Sucralose 0.2 mg 0.05-3.0%
    Flavoring agent q.s. 0.01-1.0%
    Coloring agent q.s. 0.01-1.0%
    Povidone q.s. 50 mg q.s. 100%
  • Manufacturing Process
  • Dexmedetomidine hydrochloride along with film forming polymers and other excipients are dissolved/dispersed into a pharmaceutically acceptable solvent (preferably water) and the resulting solution is then coated (spread/cast) on an inert backing layer. Dexmedetomidine hydrochloride containing polymeric layer is further dried, separated and cut into suitable sizes using appropriate die/tools and then packed as per the requirement.
  • Formulation 3: Sub-Lingual Spray
  • TABLE 3
    Composition for a tvpical Sub-lingual spray formulation
    used for sublingual delivery
    Ingredients Quantity Ranges
    Dexmedetomidine HCl 50
    (equivalent to base) micrograms
    Propylene Glycol
    10 μL 1.0-40.0%
    Alcohol 5 μL 1.0-40.0%
    Citric acid 0.2 mg   0.1-10%
    Peppermint Oil l μL 0.05-3.0%
    Purified water q.s. 100 μL q.s. 100%
  • Manufacturing Process
  • Dexmedetomidine hydrochloride along with all other excipients are mixed in a suitable order. The resulting solution/dispersion is then filled into spray canisters using appropriate tooling. They are further processed with Metered nozzles so that a specified amount of Dexmedetomidine is delivered after actuation each time.
  • Formulation 4: Sub-Lingual Liquid Drops
  • TABLE 4
    Composition for typical Sub-lingual liquid drops used for sublingual delivery
    Ingredients Quantity
    Dexmedetomidine HCl
    10 mg
    (equivalent to base)
    Normal saline q.s
    (0.9% Sodium Chloride)
  • Manufacturing Process
  • Dexmedetomidine hydrochloride ((Catalogue No. SML0956) was dissolved in Normal saline in order to yield the concentration of 1 mg/mi of the sublingual drops.
  • Example 2
  • Evaluate the effect of sublingual and intravenous administration of Dexmedetomidine hydrochloride in rat ‘resident-intruder’ model of agitation or aggression at varying dosages.
  • The resident-intruder model is an established preclinical model of aggression and agitation, and allows spontaneous and natural expression of both offensive aggression/agitation and defensive behavior in laboratory rodents in a semi natural laboratory setting. When rodents are exposed to a novel male in their home cage environment, they perceive the novel male animal as an “intruder” and demonstrate a repertoire of defensive behaviors such as ano-genital sniffing, chasing, biting and attacking (Nelson et al., ILAR Journal (2000) 41(3): 153-162).
  • Materials and Methods:
  • Animals: 12-13 week old male Wistar rats weighing 380-400 g were used as resident males. 7-8 weeks old male rats weighing 280-300 g were used as the “intruder”. Resident rats were housed with female rats for 8 days to establish territoriality. The intruder rats were housed in groups of 3 with other male rats of similar age/body weight. All animals were maintained in a controlled environment with 22±3° C. temperature, 50±20% humidity, a light/dark cycle of 12 hours each and 15-20 fresh air changes per hour and had access to food and water ad-libitum. All animal experiments were conducted in accordance with the guidelines of the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), Government of India the Association for Assessment and Accreditation of Laboratory Animal Care international (AAALAC).
  • Formulation tested: The required quantity of Formulation 4 of dexmedetomidine hydrochloride was weighed and serial dilutions were made to obtain respective doses as per the Table 5. Dilutions were prepared fresh every day prior to dosing using 0.9% normal saline from the formulation 4 for the entire study.
  • Experimental Procedure: Following acclimatization for a period of 3-5 days, each resident male rat was housed with a female rat for 8 days. On day 8, basal aggression in the resident males was tested by exposing them to an “intruder rat” for 10 minutes. Only animals that demonstrated aggression in this basal aggression test were used for the study. These animals were then randomized using body weight stratification method. The weight variation of the animals did not exceed 20% of the mean body weight in a group at the time of randomization. Animals were housed with the female rat for an additional day. On day 9, the resident animal was paired with intruder animal of an appropriate bodyweight such that the body weight of the resident was always higher than the intruder. This was to facilitate dominant, aggressive behavior in the resident animals. After randomization, animals were assigned a permanent number. Cages were identified by cage cards indicating the study number, study code, group number, sex, dose, cage number and animal number details.
  • Resident male rats were dosed with different doses of Dexmedetomidine hydrochloride (Dex) 15 minutes prior to the behavioral testing either sublingually or intravenously (Table 5). For sublingual dosing, the rats were held in one hand and using a blunt spatula the tongue was moved to one side of the mouth. Dexmedetomidine hydrochloride was then administered sublingually as liquid drops at specific concentration using a micropipette and allowed to be absorbed for a duration of 50-60 seconds. Diazepam was used as a reference compound and was dosed intraperitoneally. Vehicle controls were treated with 0.9% saline administered sublingually or intravenously. Normal controls (NC) did not received any treatment.
  • The behavior of the resident rat was recorded using an overhead video camera for 15 minutes and offline behavioral analysis was done using the Noldus Ethovision XT software. To distinguish the resident rat from the intruder rat in the video recording, the intruder rat was marked with non-toxic paint. For analysing the potential effects of Dexmedetomidine hydrochloride on agitation, we quantified various behavioral parameters such as anogenital sniffing, chasing, biting, attacking and latency to attack as well as neutral behavioral parameters such as exploration grooming, and immobile quiet time.
  • TABLE 5
    Efficacy Study: Drug treatment groups
    Cohort 1 Cohort 2
    (Sublingual dosing - (Intravenous dosing -
    Group No. of Formulation 4 adjusted to Dexmedetomidine hydrochloride
    No. animals following doses) in water or Normal saline)
    1 8 Normal Control
    2 8 Vehicle control Vehicle control
    3 8 Dexmedetomidine hydrochloride Dexmedetomidine hydrochloride
    (0.5 μg/kg) (0.5 μg/kg)
    4 8 Dexmedetomidine hydrochloride Dexmedetomidine hydrochloride
    (1.0 μg/kg) (1.0 μg/kg)
    5 8 Dexmedetomidine hydrochloride Dexmedetomidine hydrochloride
    (1.5 μg/kg) (1.5 μg/kg)
    6 8 Dexmedetomidine hydrochloride Dexmedetomidine hydrochloride
    (3.0 μg/kg) (3.0 μg/kg)
    7 8 Diazepam (3 mg/kg, i.p.)
  • Statistical Analysis: Statistical analysis was performed using validated statistical software (GraphPad Prism 6). Data is represented as Mean±SEM. One-way ANOVA (analysis of variance) followed by “Dunnett's Multiple Comparison Test” at 95% confidence interval was applied for comparison of the relevant groups. p<0.05 was considered significant.
  • Results: The present study was performed to evaluate the effect of sublingually/intravenously administered different doses of Dexmedetomidine hydrochloride on agitated behavior in a rat resident-intruder model of aggression and agitation behavior.
  • Effect of Sublingually/Intravenously Administered Dexmedetomidine Hydrochloride on Aggressive/Agitative Behavior in the Rat Resident Intruder Model:
  • The rats demonstrate a variety of defensive agitated behaviors such as anogenital sniffing, chasing, biting and attacking (indices of agitative and aggressive behavior) when exposed to a novel male in their home cage environment. The non-resident male is perceived as intruder and the resident male gets agitated and attacks the intruder male to protect their home territory. In the present experiments, vehicle treated rats demonstrated a wide repertoire of aggressive behaviors and the intruder rat was subjected to anogenital sniffing, attack, chasing and biting by the resident or dominant rat.
  • Dexmedetomidine hydrochloride (Dex) administered sublingually reduced the frequency and duration of these behaviors in a dose related manner (FIG. 1A, and FIG. 1B). Significant reduction was observed in chasing and attacking compared to vehicle control group. Similarly, intravenous administration of dexmedetomidine hydrochloride (Dex) reduced all the indices of aggressive and agitated behaviors (FIG. 1C and FIG. 1D). A significant reduction in anogenital sniffing, biting and attacking compared to vehicle controls was observed at doses above 0.5 μg/kg (FIG. 1C and FIG. 1D). Reference compound diazepam (3 mg/kg, i.p) also produced significant reduction in all the indices of aggressive and agitated behaviors evaluated in this study (FIG. 1A-1D).
  • Effect of Sublingually/Intravenously Administered Dexmedetomidine Hydrochloride on Latency to Attack
  • In addition to the change in frequency and duration of attack by the resident male, we also evaluated the effect of Dexmedetomidine hydrochloride (Dex) on the latency to attack the intruder rat. We observed an increase in the latency to attack the intruder rat following sublingual administration of Dexmedetomidine hydrochloride (Dex) in a dose related fashion indicating a reduction in aggression and agitation (FIG. 2A). When Dexmedetomidine hydrochloride (Dex) was administered intravenously, a similar increase in the latency to attack the intruder rat occurred in a dose related fashion that was significant compared to vehicle controls at a dose of 3 μg/kg (FIG. 2B). Animals treated with diazepam demonstrated a complete lack of attacking behavior (FIGS. 2A and 2B).
  • Effect of Sublingually/Intravenously Administered Dexmedetomidine Hydrochloride on Neutral Behaviors
  • Neutral behaviors like grooming, exploration and immobile/quiet time were assessed following treatment with Dexmedetomidine hydrochloride. No significant changes occurred in the grooming and exploration following sublingual administration of Dexmedetomidine hydrochloride except a reduction in exploration observed at doses of 1.5 μg/kg & 3 μg/kg (FIGS. 3A and 3B), compared to vehicle controls. Similarly, intravenously administered Dexmedetomidine hydrochloride did not significantly affect grooming and exploration in comparison to vehicle controls except at a dose of 3 μg/kg. In case of immobile/quiet time, there was no significant effect of sublingually administered Dexmedetomidine hydrochloride compared to vehicle controls however, intravenously administered Dexmedetomidine hydrochloride significantly increased the immobile/quiet time at a dose of 3 μg/kg (FIG. 3C, and FIG. 3F). Reference compound Diazepam (3 mg/kg, ip) significantly reduced the frequency and duration of all neutral behaviors evaluated in this study.
  • Interpretation
  • In the present study, we investigated the potential of Dexmedetomidine hydrochloride in reducing aggression and agitation in rat resident-intruder model. The resident-intruder model is an established preclinical model of aggression/agitation and allows spontaneous and natural expression of both offensive aggression/agitation and defensive behavior in laboratory rodents in a semi natural laboratory setting.
  • 1. Sublingual administration of Dexmedetomidine hydrochloride resulted in a dose related reduction in several behavioral indices of aggression and agitation such as anogenital sniffing, chasing, attacking and biting.
  • 2. A significant increase in the latency to attack the intruder rat was observed in a dose related manner with prior treatment with Dexmedetomidine hydrochloride as compared to the vehicle control group.
  • 3. No changes were observed in neutral behavior of animals, indicating the lack of overt anxiety-like behavior in the resident rats treated with sublingually administered Dexmedetomidine hydrochloride.
  • 4. Of the doses that were used in the study (0.5-3 μg/kg), doses of 1-1.5 μg/kg (doses administered sublingually or intravenously) effectively reduced the behavioral indices of aggression and agitation without majorly impacting the neutral behaviors.
  • Conclusion: Dexmedetomidine hydrochloride effectively reduces various indices of agitation and aggression in rat resident intruder model. Dose of 1-1.5 μg/kg effectively reduced the behavioral indices of aggression and agitation without majorly impacting the neutral behaviors. In the present study the efficacy of sublingually administered Dexmedetomidine hydrochloride correlates with intravenously administered Dexmedetomidine hydrochloride at these doses (Table 6).
  • TABLE 6
    p values obtained after statistical comparison of sublingual vs intravenous route of
    administration using Student’s t-test
    Duration (sec)
    Attack/ Anogenital- Latency
    Group Chasing Biting Fighting sniffing to attack
    NC 1.000 1.000 1.000 1.000 1.000
    Vehicle 0.207 0.069 0.290 0.753 0.136
    1 μg/kg 0.506 0.102 0.204 0.090 0.207
    1.5 μg/kg 0.125 0.059 0.107 0.727 0.508
    No significant differences (i.e. similar effect via sublingual and intravenous routes) were observed in the duration of the behavioral indices of aggression and agitation (chasing, biting, attack, anogenital sniffing latency to attack) when compared between sublingual and intravenous routes of dexmedetomidine hydrochloride administration at doses of 1 and 1.5 μg/kg. Statistical analysis was performed using student t-test. *p < 0.05, **p < 0.01 ***p < 0.001 and ****p < 0.0001 Sublingual vs intravenous routes of administration.
  • Based on 1-1.5 μg/kg rat efficacy doses, the human equivalent sublingual doses are calculated to be 0.161 μg/kg & 0.242 μg/kg. The total human equivalent dose for a 60-kg human would be 10 and 15 μg (https://www.fda.gov/downloads/drugs/guidances/ucm078932.pdf).
  • Example 3: Estimation of Dexmedetomidine (0.5-3 μg/Kg) in Rat Plasma Samples by LC-MS/MS
  • Objective: To estimate Dexmedetomidine levels in rat plasma samples obtained after dosing animals via intravenous and sublingual routes at doses of 0.5, 1, 1.5 and 3 μg/kg.
  • Blood collection: To determine the plasma concentration of dexmedetomidine, Dexmedetomidine hydrochloride was administered sublingually or intravenously in rats (n=3) at different doses (Formulation 4 adjusted to 0.5, 1, 1.5, 3 μg/kg). Blood was collected under mild isoflurane anesthesia from the retro-orbital plexus at 0, 5, 15, 30, 60 and 120 minutes post dosing. Plasma was separated and stored at −80° C. until Dexmedetomidine concentration was analyzed.
  • Materials and Methods
  • Preparation of Standard Solutions
  • A standard stock solution of dexmedetomidine hydrochloride was prepared by dissolving 1.358 mg of dexmedetomidine hydrochloride in 1358 μl of milli-Q water to achieve a concentration of 829.071 mg/ml. Working solutions of different concentrations were prepared by using diluent (methanol:water (50:50) % v/v).
  • Tolbutamide was used as an internal standard and its stock solution was prepared by dissolving 25 mg of tolbutamide in 1000 μl of DMSO to achieve a concentration of 25 mg/ml. Working solutions of different concentrations were prepared by using a diluent (acetonitrile: water (50:50) % v/v).
  • Solution preparation for SPE and chromatography: Mobile phase A (10 mm ammonium formate, pH 3.50): 0.6306 gms of ammonium formate was weighed and transferred to a 1000 ml reagent bottle. To this, 1000 ml of milli q water was added and pH of the resulting solution was adjusted to 3.5 using formic acid.
  • Mobile phase B: 100% acetonitrile
  • Diluent (methanol:water (50:50) % v/v): 50 ml of methanol was mixed with 50 ml of milli-q water. Resulting solution was used as diluent.
  • Wash solution: 100 μl of ammonia was mixed with 100 ml of milli q. Resulting solution was used as wash solution.
  • Elution solvent: 100 μl of formic acid was mixed with 100 ml of acetonitrile. Resulting solution was used as elution solvent.
  • Analytical Methods: Samples were analysed by using Agilent 1290 Infinity II HPLC system coupled to AB Sciex Triple Quad instrument (API-5000). Chromatographic separation was done using Agilent Zorbax Eclipse plus C18 column (50*2.1 mm, 1.8 μm) in gradient mode. The mobile phase consisted of 10 mM Ammonium Formate with pH 3.5 (Mobile phase A) and 100% Acetonitrile (Mobile phase B). The column temperature was 40° C. and flow rate was 0.35 mL/min. The MS instrument was operated in the positive mode (ESI+). For analysis, 2 μL of sample was injected into the LC-MS/MS instrument. Auto sampler temperature was 7° C.
  • Quality control (QC) samples were prepared as following as per table 7:
  • TABLE 7
    Dexmedetomidine Volume Final
    conc of Blank Total Calibration
    (Solution A) solution plasma Volume Conc QC
    (ng/mL) A (μL) (μL) (μL) (pg/mL) ID
    1.114 2 48 50 44.571 LQC
    371.424 2 48 50 14856.962 MQC
    928.560 2 48 50 37142.406 HQC
  • Sample Preparation
  • WCX SPE 96 well plate was used for sample preparation. 50 μl of plasma sample was used for extraction. Along with study samples, one set of linearity and two sets of quality controls (QC) were also processed.
  • Sample pretreatment: To 50 μl of plasma, 10 μl of tolbutamide working solution was added (Tolbutamide 250 ng/ml). After mixing, 50 μL of buffer solution (10 mM Ammonium Formate pH 3.5) was added. Contents were vortex mixed and loaded to preconditioned SPE plate.
  • LC-MS/MS Analysis
  • After placing the cartridges in the negative pressure SPE unit, they were conditioned by passing 200 μl of 100% methanol followed by 200 μl of water. The pretreated plasma samples were then loaded to the pre-conditioned cartridges.
  • After loading pretreated plasma samples, cartridges were washed with 100 μl of 0.1% ammonia solution. Finally, bound analyte was eluted with 50 μl of 0.1% formic acid in acetonitrile. This step was repeated twice for complete elution. Final eluent volume was 100 μL. To 100 μL of eluent, 50 μL of 10 mM ammonium formate (pH 3.5) was added samples were vortex mixed and transferred to a 96-well HPLC sample plate (Agilent) and submitted for LC-MS/MS analysis. For LC-MS/MS analysis, 2 μL of sample was injected. Calibration standards and QCs were processed the same way as done for study samples.
  • Mean plasma concentrations of Dexmedetomidine in various rat plasma samples at various time points was determined by LC-MS/MS method using Analyst 1.6.2 software (Table 8 and FIGS. 4A and 4B) with a calibration curve in the range of 0.011-53.061 ng/ml prepared in blank rat plasma matrix. The calibration curve was fitted by linear regression. The concentrations in the QC and test samples (pg/mL) were obtained from the Analyst software based on the calibration curve. Acceptance criteria for the calibration curve and QCs are as follows: 1) At least 75% of the non-zero calibration standards must be included in the calibration curve with all back-calculated concentrations within ±20% deviation from nominal concentrations (except for the lower level of quantification, LLOQ, where ±20% deviation is acceptable). 2) The correlation coefficient (r) of the calibration curve must be greater than or equal to 0.99. 3) At least two-thirds (4 out of 6) QC samples must be within ±20% relative error (accuracy)
  • Results:
  • TABLE 8
    Mean rat plasma concentrations following Sublingual or Intravenous
    dexmedetomidine hydrochloride administration at varying doses
    Sublingual Mean Concentration in pg/mL at Intravenous Concentration in pg/mL at
    Dosing various time points after dosing dosing various time points after dosing
    groups
    0 5 15 30 60 120 groups 0 5 15 30 60 120
    (I-IV) min min min min min min (V-VIII) min min min min min min
    I- BLQ 48 ± 51 ± 87 ± 17 ± BLQ V- BLQ 70 ± 46 ± 35 ± 19 ± BLQ
    Dex. HCl 30.4 29.1 89.7 0.7 Dex. HCl 7.2 14.2 4.9 3.5
    0.5 μg/kg, 0.5 μg/kg,
    SL i.v.
    II- BLQ 51 ± 47 ± 43 ± 13 ± 19 ± VI- BLQ 174 ± 90 ± 45 ± 63 ± BLQ
    Dex. HCl 44.7 22.4 13.5 2.8 7.07 Dex. HCl 12.5 12.1 1.7 58.0
    1 μg/kg, 1 μg/kg,
    SL i.v.
    III- BLQ 84 ± 27 ± 31 ± 37 ± 36 ± VII- BLQ 158 ± 114 ± 65 ± 31 ± 21 ±
    Dex. HCl 37.7 7.1 5.5 16.3 6.36 Dex. HCl 56.1 1.7 11.0 10.3 8.89
    1.5 μg/kg, 1.5 μg/kg,
    SL i.v.
    IV- BLQ 71 ± 42 ± 160 ± 96 ± 93 ± VIII- BLQ 471 ± 266 ± 139 ± 84 ± 34 ±
    Dex. HCl 52.0 13.0 117.9 21.5 53.95 Dex. HCl 24.9 31.6 18.0 17.4 9.61
    3 μg/kg, 3 μg/kg,
    SL i.v.
    BLQ: Below the Lowest limit of Quantification of the assay (LOQ: 0.05 ng/ml)
    SL: sublingual;
    i.v.: intravenous
    Data expressed as Mean ± SD
  • INTERPRETATION AND CONCLUSION
  • Following sublingual administration of Dexmedetomidine hydrochloride, a dose-related effect on plasma concentrations was observed at doses ranging from 0.5-3 μg/kg (FIG. 4A, table 8).
  • Following intravenous administration of Dexmedetomidine hydrochloride, a dose-dependent effect on plasma concentrations was observed at doses ranging from 0.5-3 μg/kg (FIG. 4B, table 8).
  • Doses of 1 and 1.5 μg/kg effectively reduced various indices of agitation and aggression without majorly impacting neutral behaviors. Plasma concentrations following administration of dose of 1 μg/kg (via sublingual and intravenous route) between 15 to 30 min (time corresponding to the time of behavioral response observed in the efficacy study; drug administered 15 min prior to agitation behavior test & animal observed for 15 min) range from 43±13.5 to 90±12.1 μg/ml (Table 8). Similarly, plasma concentrations following administration of dose of 1.5 μg/kg (via sublingual and intravenous route) between 15 to 30 min range from 27±7.1-114±1.7 pg/ml (Table 8).

Claims (17)

What is claimed is:
1. A method of treating agitation in an agitated human subject, said agitated human subject having schizophrenia or bipolar disorder, comprising administering oromucosally to said agitated human subject a dissolvable film comprising an effective amount of dexmedetomidine or a pharmaceutically acceptable salt thereof, and wherein said dexmedetomidine or a pharmaceutically acceptable salt thereof is administered as a single dosage.
2. The method of claim 1, wherein said dexmedetomidine or a pharmaceutically acceptable salt thereof is administered sublingually.
3. The method of claim 1, wherein said dexmedetomidine or a pharmaceutically acceptable salt thereof is administered buccally.
4. The method of claim 1, wherein said dexmedetomidine or a pharmaceutically acceptable salt thereof is dexmedetomidine hydrochloride.
5. The method of claim 1, wherein said agitated human subject has schizophrenia.
6. The method of claim 1, wherein said agitated human subject has bipolar disorder.
7. The method of claim 1, wherein said single dosage is about 3 micrograms to about 100 micrograms.
8. The method of claim 7, wherein said single dosage is about 5 micrograms to about 90 micrograms.
9. The method of claim 7, wherein said single dosage is about 5 micrograms to about 80 micrograms.
10. The method of claim 7, wherein said single dosage is about 5 micrograms to about 70 micrograms.
11. The method of claim 7, wherein said single dosage is about 5 micrograms to about 60 micrograms.
12. The method of claim 7, wherein said single dosage is about 5 micrograms to about 40 micrograms.
13. The method of claim 7, wherein said single dosage is about 30 micrograms.
14. The method of claim 7, wherein said single dosage is about 60 micrograms.
15. The method of claim 7, wherein said single dosage is about 90 micrograms.
16. The method of claim 1, wherein the agitation is acute agitation.
17. The method of claim 1, wherein the agitation is chronic agitation.
US17/990,332 2016-12-31 2022-11-18 Use of sublingual dexmedetomidine for the treatment of agitation Pending US20230093109A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/990,332 US20230093109A1 (en) 2016-12-31 2022-11-18 Use of sublingual dexmedetomidine for the treatment of agitation

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201662441164P 2016-12-31 2016-12-31
US201762471393P 2017-03-15 2017-03-15
US201762542323P 2017-08-08 2017-08-08
PCT/US2017/069030 WO2018126182A1 (en) 2016-12-31 2017-12-29 Use of sublingual dexmedetomidine for the treatment of agitation
US201916474882A 2019-06-28 2019-06-28
US17/990,332 US20230093109A1 (en) 2016-12-31 2022-11-18 Use of sublingual dexmedetomidine for the treatment of agitation

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US16/474,882 Continuation US11839604B2 (en) 2016-12-31 2017-12-29 Use of sublingual dexmedetomidine for the treatment of agitation
PCT/US2017/069030 Continuation WO2018126182A1 (en) 2016-12-31 2017-12-29 Use of sublingual dexmedetomidine for the treatment of agitation

Publications (1)

Publication Number Publication Date
US20230093109A1 true US20230093109A1 (en) 2023-03-23

Family

ID=62710955

Family Applications (5)

Application Number Title Priority Date Filing Date
US16/474,882 Active US11839604B2 (en) 2016-12-31 2017-12-29 Use of sublingual dexmedetomidine for the treatment of agitation
US17/496,470 Active US11931340B2 (en) 2016-12-31 2021-10-07 Use of sublingual dexmedetomidine for the treatment of agitation
US17/990,312 Active US11786508B2 (en) 2016-12-31 2022-11-18 Use of sublingual dexmedetomidine for the treatment of agitation
US17/990,332 Pending US20230093109A1 (en) 2016-12-31 2022-11-18 Use of sublingual dexmedetomidine for the treatment of agitation
US18/124,943 Pending US20230218580A1 (en) 2016-12-31 2023-03-22 Use of sublingual dexmedetomidine for the treatment of agitation

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US16/474,882 Active US11839604B2 (en) 2016-12-31 2017-12-29 Use of sublingual dexmedetomidine for the treatment of agitation
US17/496,470 Active US11931340B2 (en) 2016-12-31 2021-10-07 Use of sublingual dexmedetomidine for the treatment of agitation
US17/990,312 Active US11786508B2 (en) 2016-12-31 2022-11-18 Use of sublingual dexmedetomidine for the treatment of agitation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/124,943 Pending US20230218580A1 (en) 2016-12-31 2023-03-22 Use of sublingual dexmedetomidine for the treatment of agitation

Country Status (14)

Country Link
US (5) US11839604B2 (en)
EP (1) EP3562486B1 (en)
JP (5) JP6868698B2 (en)
KR (2) KR20240031326A (en)
CN (1) CN110337290A (en)
AU (2) AU2017388759B2 (en)
BR (1) BR112019013503A2 (en)
CA (1) CA3045043A1 (en)
IL (1) IL267689A (en)
MX (3) MX2019007923A (en)
SG (1) SG10202107367SA (en)
TW (1) TWI791479B (en)
WO (1) WO2018126182A1 (en)
ZA (1) ZA202006636B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11786508B2 (en) 2016-12-31 2023-10-17 Bioxcel Therapeutics, Inc. Use of sublingual dexmedetomidine for the treatment of agitation
US11806429B2 (en) 2018-06-27 2023-11-07 Bioxcel Therapeutics, Inc. Film formulations containing dexmedetomidine and methods of producing them
US11890272B2 (en) 2019-07-19 2024-02-06 Bioxcel Therapeutics, Inc. Non-sedating dexmedetomidine treatment regimens

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210267944A1 (en) * 2018-06-27 2021-09-02 Bioxcel Therapeutics, Inc. Methods for treating agitation using dexmedetomidine hydrochloride
RU2692247C1 (en) * 2018-12-06 2019-06-24 федеральное государственное бюджетное учреждение "Национальный медицинский исследовательский центр имени В.А. Алмазова" Министерства здравоохранения Российской Федерации Method of postoperative delirium treatment
CA3154659A1 (en) * 2019-09-18 2021-03-25 Bioxcel Therapeutics, Inc. Systems and methods for detection and prevention of emergence of agitation
KR102398219B1 (en) * 2020-02-07 2022-05-17 주식회사 뉴로벤티 Composition comprising rilmenidine compound for treating fragile x syndrome and related developmental disorders
EP4225305A1 (en) * 2020-10-08 2023-08-16 Bioxcel Therapeutics, Inc. Treatment of bipolar disorders and psychosis using dexmedetomidine hydrochloride
KR20230159515A (en) 2021-03-19 2023-11-21 오리온 코포레이션 Tacipimidine formulations and uses thereof
GB2618810A (en) * 2022-05-17 2023-11-22 Novumgen Ltd Pharmaceutical composition for sublingual administration of clonidine
WO2024023261A1 (en) 2022-07-27 2024-02-01 Universität Zürich Dexmedetomidine for the treatment of sleep disorders
WO2024055042A1 (en) * 2022-09-11 2024-03-14 Bioxcel Therapeutics, Inc. Methods for treating agitation in community settings
US11806334B1 (en) 2023-01-12 2023-11-07 Bioxcel Therapeutics, Inc. Non-sedating dexmedetomidine treatment regimens

Family Cites Families (258)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU1138164A1 (en) 1963-04-25 1985-02-07 Shadurskij Konstantin S Antihistamine agent "dimebon"
US4407957A (en) 1981-03-13 1983-10-04 Damon Corporation Reversible microencapsulation of a core material
FI844786A0 (en) 1984-12-04 1984-12-04 Farmos Oy TERAPEUTISKT UTNYTTJBAR FOERENING.
US4839170A (en) 1985-10-01 1989-06-13 Survival Technology, Inc. Protein absorption enhancing agents
US4767789A (en) 1986-10-21 1988-08-30 American Home Products Corporation (Del.) Spray dried acetaminophen
US4760093A (en) 1986-10-21 1988-07-26 American Home Products Corporation (Del.) Spray dried acetaminophen
US4760094A (en) 1986-10-21 1988-07-26 American Home Products Corporation (Del.) Spray dried acetaminophen
US5039540A (en) 1989-08-14 1991-08-13 Neophore Technologies, Inc. Freeze dry composition and method for oral administration of drugs, biologicals, nutrients and foodstuffs
US5217718A (en) 1989-08-18 1993-06-08 Cygnus Therapeutic Systems Method and device for administering dexmedetomidine transdermally
US5178878A (en) 1989-10-02 1993-01-12 Cima Labs, Inc. Effervescent dosage form with microparticles
US5215756A (en) 1989-12-22 1993-06-01 Gole Dilip J Preparation of pharmaceutical and other matrix systems by solid-state dissolution
US5188825A (en) 1989-12-28 1993-02-23 Iles Martin C Freeze-dried dosage forms and methods for preparing the same
EP0585368B1 (en) 1991-04-25 1997-08-06 Brown University Research Foundation Implantable biocompatible immunoisolatory vehicle for delivery of selected therapeutic products
US5352456A (en) 1991-10-10 1994-10-04 Cygnus Therapeutic Systems Device for administering drug transdermally which provides an initial pulse of drug
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
DE69332105T2 (en) 1992-09-29 2003-03-06 Inhale Therapeutic Systems San PULMONAL DELIVERY OF ACTIVE FRAGMENT OF PARATHORMON
US5343672A (en) 1992-12-01 1994-09-06 Scherer Ltd R P Method for manufacturing freeze dried dosages in a multilaminate blister pack
ATE208615T1 (en) 1993-07-09 2001-11-15 Scherer Corp R P METHOD FOR PRODUCING FREEZE-DRIED MEDICINAL DOSAGE FORMS
GB2281206A (en) 1993-08-25 1995-03-01 Orion Yhtymae Oy Use of dexmedetomidine
US5457895A (en) 1993-10-01 1995-10-17 R. P. Scherer Corporation Method of identifying freeze-dried dosage forms
US5508367A (en) 1993-11-29 1996-04-16 Adhesives Research, Inc. Water-soluble pressure sensitive adhesive
US5395907A (en) 1993-11-29 1995-03-07 Adhesive Research, Inc. Water-soluble pressure sensitive adhesive
EP0681601B1 (en) 1993-11-29 1999-02-17 Adhesives Research, Inc. Water-soluble pressure sensitive adhesive
FR2718357B1 (en) 1994-04-06 1997-10-03 Defarges Alain Moreau Improvements made to a needleless jet injection device.
GB2290964A (en) 1994-07-08 1996-01-17 Arto Olavi Urtti Transdermal drug delivery system
US5731387A (en) 1994-07-11 1998-03-24 Adhesives Research, Inc. Ionically-crosslinked water-absorbent graft copolymer
US5726250A (en) 1994-07-11 1998-03-10 Adhesives Research, Inc. Covalently crosslinked water-absorbent graft copolymer
US5837287A (en) 1994-10-28 1998-11-17 R P Scherer Corporation Process for preparing solid pharmaceutical dosage forms
US6726928B2 (en) 1994-10-28 2004-04-27 R.P. Scherer Technologies, Inc. Process for preparing solid dosage forms for unpalatable pharmaceuticals
GB9421836D0 (en) 1994-10-28 1994-12-14 Scherer Corp R P Process for preparing solid pharmaceutical dosage forms of hydrophobic substances
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
US5605911A (en) 1995-01-31 1997-02-25 Washington University Use of alpha-2 adrenergic drugs to prevent adverse effects of NMDA receptor hypofunction (NRH)
US5700873A (en) 1995-03-07 1997-12-23 Adhesives Research, Inc. Method of preparation of water-soluble copolymer
US5780014A (en) 1995-04-14 1998-07-14 Inhale Therapeutic Systems Method and apparatus for pulmonary administration of dry powder alpha 1-antitrypsin
US5654007A (en) 1995-06-07 1997-08-05 Inhale Therapeutic Systems Methods and system for processing dispersible fine powders
GB9616672D0 (en) 1996-08-08 1996-09-25 Scherer Ltd R P Pharmaceutical compositions
US5800832A (en) 1996-10-18 1998-09-01 Virotex Corporation Bioerodable film for delivery of pharmaceutical compounds to mucosal surfaces
GB9700624D0 (en) 1997-01-14 1997-03-05 Danbiosyst Uk Drug delivery composition
GB9702799D0 (en) 1997-02-12 1997-04-02 Scherer Corp R P Process for preparing solid pharmaceutical dosage forms
AU736912B2 (en) 1997-02-20 2001-08-02 Therics, Inc. Dosage form exhibiting rapid disperse properties, methods of use and process for the manufacture of same
US5951999A (en) 1997-02-21 1999-09-14 Adhesives Research, Inc. Transdermal pressure sensitive adhesive drug delivery system
US6239228B1 (en) 1997-02-21 2001-05-29 Adhesives Research, Inc. Pressure sensitive adhesive containing macromer having repeat hydrophilic moieties
KR20000070375A (en) 1997-02-21 2000-11-25 에드워드 엘. 데이지 Transdermal pressure sensitive adhesive drug delivery system and pressure sensitive adhesive used therein
US6024981A (en) 1997-04-16 2000-02-15 Cima Labs Inc. Rapidly dissolving robust dosage form
US5976577A (en) 1997-07-11 1999-11-02 Rp Scherer Corporation Process for preparing fast dispersing solid oral dosage form
EP1001748B1 (en) 1997-07-25 2006-04-19 Alpex Pharma S.A. A process for the preparation of a granulate suitable to the preparation of rapidly disintegrable mouth-soluble tablets
US7632517B2 (en) 1997-10-01 2009-12-15 Novadel Pharma Inc. Buccal, polar and non-polar spray containing zolpidem
GB9802088D0 (en) 1998-01-30 1998-03-25 Scherer Ltd R P Pharmaceutical products
JP4237405B2 (en) 1998-03-09 2009-03-11 シーマ・ラブス、インコーポレイテッド Assembly
US6200604B1 (en) 1998-03-27 2001-03-13 Cima Labs Inc. Sublingual buccal effervescent
US6716867B1 (en) 1998-04-01 2004-04-06 Orion Corporation Use of dexmedetomidine for ICU sedation
AU2005201500B2 (en) 1998-06-03 2007-01-04 Scott Laboratories, Inc. Apparatus and method for providing a conscious patient relief from pain and anxiety associated with medical or surgical procedures
US7565905B2 (en) 1998-06-03 2009-07-28 Scott Laboratories, Inc. Apparatuses and methods for automatically assessing and monitoring a patient's responsiveness
US6807965B1 (en) 1998-06-03 2004-10-26 Scott Laboratories, Inc. Apparatus and method for providing a conscious patient relief from pain and anxiety associated with medical or surgical procedures
US7001609B1 (en) 1998-10-02 2006-02-21 Regents Of The University Of Minnesota Mucosal originated drug delivery systems and animal applications
ID29270A (en) 1998-11-20 2001-08-16 Rtp Pharma Inc MICRO PARTICLES THAT ARE STABILIZED BY PHOSPHOLIPIDS THAT CAN SPREAD
GB9901819D0 (en) 1999-01-27 1999-03-17 Scherer Corp R P Pharmaceutical compositions
GB9908014D0 (en) 1999-04-08 1999-06-02 Scherer Corp R P Pharmaceutical compositions
US6284270B1 (en) 1999-08-04 2001-09-04 Drugtech Corporation Means for creating a mass having structural integrity
DE19940740A1 (en) 1999-08-31 2001-03-01 Gruenenthal Gmbh Pharmaceutical salts
US6984207B1 (en) 1999-09-14 2006-01-10 Hoana Medical, Inc. Passive physiological monitoring (P2M) system
CA2324967A1 (en) 2000-11-01 2002-05-01 3816133 Canada Inc. System for monitoring patients with alzheimer's disease or related dementia
US6316029B1 (en) 2000-05-18 2001-11-13 Flak Pharma International, Ltd. Rapidly disintegrating solid oral dosage form
AU7323001A (en) 2000-07-07 2002-01-21 Av Topchiev Inst Petrochemical Preparation of hydrophilic pressure sensitive adhesives having optimized adhesive properties
US6982251B2 (en) 2000-12-20 2006-01-03 Schering Corporation Substituted 2-azetidinones useful as hypocholesterolemic agents
US6814978B2 (en) 2000-12-29 2004-11-09 Mcneil-Ppc, Inc. Process for preparing a soft tablet
WO2002089794A1 (en) 2001-05-07 2002-11-14 Universite Catholique De Louvain Method for treating neuropathic pain and pharmaceutical preparation therefor
US20030083822A2 (en) 2001-05-15 2003-05-01 Psychogenics, Inc. Systems and methods for monitoring behavior informatics
US6509040B1 (en) 2001-06-22 2003-01-21 R.P. Scherer Corporation Fast dispersing dosage forms essentially free of mammalian gelatin
US8663687B2 (en) 2001-10-12 2014-03-04 Monosol Rx, Llc Film compositions for delivery of actives
US8603514B2 (en) 2002-04-11 2013-12-10 Monosol Rx, Llc Uniform films for rapid dissolve dosage form incorporating taste-masking compositions
US20110033542A1 (en) 2009-08-07 2011-02-10 Monosol Rx, Llc Sublingual and buccal film compositions
JP2005508939A (en) 2001-10-12 2005-04-07 エラン ファーマ インターナショナル,リミティド Composition having combined immediate release and sustained release characteristics
US7425292B2 (en) 2001-10-12 2008-09-16 Monosol Rx, Llc Thin film with non-self-aggregating uniform heterogeneity and drug delivery systems made therefrom
US20070281003A1 (en) 2001-10-12 2007-12-06 Fuisz Richard C Polymer-Based Films and Drug Delivery Systems Made Therefrom
US20140271788A1 (en) 2013-03-15 2014-09-18 Monosol Rx, Llc Sublingual and buccal film compositions
US7666337B2 (en) 2002-04-11 2010-02-23 Monosol Rx, Llc Polyethylene oxide-based films and drug delivery systems made therefrom
US8900498B2 (en) 2001-10-12 2014-12-02 Monosol Rx, Llc Process for manufacturing a resulting multi-layer pharmaceutical film
US7357891B2 (en) 2001-10-12 2008-04-15 Monosol Rx, Llc Process for making an ingestible film
US8765167B2 (en) 2001-10-12 2014-07-01 Monosol Rx, Llc Uniform films for rapid-dissolve dosage form incorporating anti-tacking compositions
FR2832311B1 (en) 2001-11-21 2004-04-16 Besins Int Belgique FILM-FORMING POWDER, COMPOSITIONS COMPRISING SAME, PREPARATION METHODS AND USES THEREOF
FR2834212B1 (en) 2001-12-27 2004-07-09 Besins Int Belgique USE OF IMMEDIATE RELEASE POWDER IN PHARMACEUTICAL AND NUTRACEUTICAL COMPOSITIONS
EP1485296B1 (en) 2002-02-13 2011-07-20 Michael K. Weibel Drug dose - form and method of manufacture
EP1549305B1 (en) 2002-10-08 2009-04-22 Allergan, Inc. Alpha 2b 0r 2b/2c adrenoceptor agonists for the treatment of morbus alzheimer and morbus parkinson
US20040156894A1 (en) 2003-02-07 2004-08-12 Grother Leon Paul Use of edible acids in fast-dispersing pharmaceutical solid dosage forms
EP1663178A1 (en) 2003-05-28 2006-06-07 MonoSolRX, LLC Films based on poly(ethylene oxide), and drug delivery systems made thereof
US7282217B1 (en) 2003-08-29 2007-10-16 Kv Pharmaceutical Company Rapidly disintegrable tablets
US9248146B2 (en) 2003-10-24 2016-02-02 Adhesives Research, Inc. Dissolvable adhesive films for delivery of pharmaceutical or cosmetic agents
EP1680079A4 (en) 2003-10-24 2008-01-02 Adhesives Res Inc Rapidly disintegrating films for delivery of pharmaceutical or cosmetic agents
GB0403808D0 (en) 2004-02-20 2004-03-24 Bioprogress Technology Ltd Films for use as dosage forms
US20050222270A1 (en) 2004-02-26 2005-10-06 Olney John W Prolonged administration of NMDA antagonist drug and safener drug to create improved stable neural homeostasis
US7972621B2 (en) 2004-06-03 2011-07-05 R.P. Scherer Technologies, Llc Process for formulating fast dispersing dosage forms comprising at least one fish gelatin selected on the basis of molecular weight
US20060058590A1 (en) 2004-08-24 2006-03-16 Shaw Geoffrey M Method and system for assaying agitation
US20060058700A1 (en) 2004-08-26 2006-03-16 Marro Dominic P Patient sedation monitor
AU2005286733B2 (en) 2004-09-23 2009-11-05 Alexander Michalow Methods for regulating neurotransmitter systems by inducing counteradaptations
EP1871219A4 (en) 2005-02-22 2011-06-01 Health Smart Ltd Methods and systems for physiological and psycho-physiological monitoring and uses thereof
US20060199805A1 (en) 2005-03-04 2006-09-07 Boehringer Ingelheim International Gmbh Pharmaceutical compositions for the treatment and/or prevention of anxiety disorders
US20090076156A1 (en) 2005-07-18 2009-03-19 Masud Husain Use of a noradrenergic agonist, e.g. guanfacine, for the treatment of cognitive disorders
US20070106126A1 (en) 2005-09-30 2007-05-10 Mannheimer Paul D Patient monitoring alarm escalation system and method
WO2007041697A2 (en) 2005-10-04 2007-04-12 Medivation, Inc. Hydrogenated pyrido-indole compounds for the treatment of huntington ' s disease
US8158152B2 (en) 2005-11-18 2012-04-17 Scidose Llc Lyophilization process and products obtained thereby
BRPI0620578A2 (en) 2005-12-27 2011-12-06 Jubilant Organosys Ltd pharmaceutical composition that dissolves in the mouth and process for the preparation thereof
PT1986642E (en) 2006-02-13 2014-01-30 Orient Pharma Samoa Co Ltd Combination of alpha-2 receptor agonist (clonidine) and an anti-muscarinic agent (oxybutynin) for the treatment of sialorrhoea
US7630758B2 (en) 2006-06-22 2009-12-08 General Electric Company Separation of natural and drug-induced sleep of a subject
US20080026040A1 (en) 2006-07-31 2008-01-31 Isaac Farr Active agent-releasing dosage forms
EP2063864A4 (en) 2006-09-20 2012-03-14 Monosol Rx Llc Edible water-soluble film containing a foam reducing flavoring agent
US8157730B2 (en) 2006-12-19 2012-04-17 Valencell, Inc. Physiological and environmental monitoring systems and methods
US20080153808A1 (en) 2006-12-22 2008-06-26 Allergan, Inc. Alpha-2 receptor pan agonist and serotonin-norepinephrine reuptake inhibitor compositions for treating chronic pain
WO2008091588A1 (en) 2007-01-22 2008-07-31 Targacept, Inc. Intranasal, buccal, and sublingual administration of metanicotine analogs
US8568777B2 (en) 2007-03-30 2013-10-29 Monosol Rx, Llc Packaged film dosage unit containing a complexate
CN101801346A (en) 2007-05-10 2010-08-11 诺瓦德尔药品公司 anti-insomnia compositions and methods
US20090099480A1 (en) 2007-05-24 2009-04-16 Peter Salgo System and method for patient monitoring
CN101686942B (en) 2007-06-27 2012-09-26 韩美药品株式会社 Method for preparing rapidly disintegrating formulation for oral administration and apparatus for preparing and packing the same
RU2338533C1 (en) 2007-06-28 2008-11-20 Сергей Олегович Бачурин AGENT POSSESSING ANXIOLYTHIC ACTION, ON BASIS OF HYDROGENATED PYRIDO (4,3-b) INDOLES (VERSIONS), PHARMACOLOGICAL AGENT ON ITS BASIS AND WAY OF ITS APPLICATION
WO2009017836A1 (en) 2007-08-01 2009-02-05 Medivation Neurology, Inc. Methods and compositions for treating schizophrenia using antipsychotic combination therapy
MX2010003928A (en) 2007-10-11 2010-09-10 Philip Morris Prod Smokeless tobacco product.
US8298583B2 (en) 2007-10-19 2012-10-30 Monosol Rx, Llc Film delivery system for tetrahydrolipstatin
PE20091084A1 (en) 2007-12-07 2009-07-23 Schering Plough Healthcare PHARMACEUTICAL FORMULATIONS OF PHENYLPHRINE AND COMPOSITIONS FOR TRANSMUCOSAL ABSORPTION
CN101496801A (en) 2008-02-02 2009-08-05 四川百利药业有限责任公司 Dexmedetomidine and use of pharmaceutical salt thereof
EP2252290B1 (en) 2008-02-15 2017-11-29 Bone Therapeutics S.A. Pharmaceutical composition for use in the treatment and/or the prevention of osteoarticular diseases
JP5547096B2 (en) 2008-02-28 2014-07-09 アール.ピー. シェーラー テクノロジーズ エルエルシー How to minimize polymorphism
US20190216389A1 (en) 2008-04-07 2019-07-18 Christopher Scheib Method and system for analyzing a series of electroencephalogram (eeg) signals during altered brain states
US20090275853A1 (en) 2008-04-30 2009-11-05 The General Electric Company Method and apparatus for monitoring physiological state of a subject
US8882684B2 (en) 2008-05-12 2014-11-11 Earlysense Ltd. Monitoring, predicting and treating clinical episodes
US20100029706A1 (en) 2008-07-30 2010-02-04 Edison Parmaceuticals, Inc. a Delaware Corporation HYDROGENATED PYRIDO[4,3-b]INDOLES FOR THE TREATMENT OF OXIDATIVE STRESS
JP2012502915A (en) 2008-09-15 2012-02-02 バイオビスタ インコーポレイテッド Compositions and methods for treating epilepsy
US8256233B2 (en) 2008-10-31 2012-09-04 The Invention Science Fund I, Llc Systems, devices, and methods for making or administering frozen particles
US8731842B2 (en) 2008-10-31 2014-05-20 The Invention Science Fund I, Llc Compositions and methods for biological remodeling with frozen particle compositions
US8470546B2 (en) 2008-11-25 2013-06-25 Albert Einstein College Of Medicine Of Yeshiva University Treatment of autism spectrum disorders with agents that activate the Locus Coeruleus-Noradrenergic system
EP2370136A4 (en) 2008-12-01 2015-12-30 Map Pharmaceuticals Inc Inhalation delivery methods and devices
US8282954B2 (en) 2008-12-15 2012-10-09 Monosol Rx, Llc Method for manufacturing edible film
TR200903014A1 (en) 2009-04-17 2010-11-22 Sanovel İlaç San. Ve Ti̇c. A.Ş. Orally dispersed dimebolin compounds.
EP2429521B1 (en) * 2009-05-15 2017-10-18 Recro Pharma, Inc. Sublingual dexmedetomidine compositions and methods of use thereof
US8475832B2 (en) 2009-08-07 2013-07-02 Rb Pharmaceuticals Limited Sublingual and buccal film compositions
US9775819B2 (en) 2009-09-16 2017-10-03 R.P. Scherer Technologies, Llc Oral solid dosage form containing nanoparticles and process of formulating the same using fish gelatin
US8313768B2 (en) 2009-09-24 2012-11-20 Mcneil-Ppc, Inc. Manufacture of tablet having immediate release region and sustained release region
WO2011039686A1 (en) 2009-09-30 2011-04-07 Pfizer Inc. Latrepirdine oral sustained release dosage forms
RU2012133969A (en) 2010-01-08 2014-02-20 Рекро Фарма, Инк. LOCAL TRANSDERMAL DEXMEDETOMIDINE COMPOSITIONS AND WAYS OF THEIR APPLICATION
WO2011109716A2 (en) 2010-03-04 2011-09-09 Neumitra LLC Devices and methods for treating psychological disorders
US10548839B2 (en) 2010-03-16 2020-02-04 Wei Tian Process of manufacturing a lyophilized fast dissolving, multi-phasic dosage form
US20130029989A1 (en) 2010-04-15 2013-01-31 The Royal Institution For The Advancement Of Learning/Mcgill University Topical treatments for pain
US9572773B2 (en) 2010-04-26 2017-02-21 Novartis A.G. Layered drug delivery device
US20110290694A1 (en) 2010-05-27 2011-12-01 Monosol Rx, Llc Oral film dosage form having indicia thereon
US8529914B2 (en) 2010-06-28 2013-09-10 Richard C. Fuisz Bioactive dose having containing a material for modulating pH of a bodily fluid to help or hinder absorption of a bioactive
US10137245B2 (en) 2010-08-17 2018-11-27 University Of Florida Research Foundation, Inc. Central site photoplethysmography, medication administration, and safety
CA2815568C (en) 2010-09-23 2018-10-09 Monosolrx Llc Method and system for forming a pharmaceutical product directly onto a packaging surface
US8383135B2 (en) 2010-12-03 2013-02-26 Richard C. Fuisz Solid dosage form that promotes reliable oral, esophageal and GI transit
KR101890317B1 (en) 2010-12-16 2018-08-22 선오비온 파마슈티컬스 인코포레이티드 Sublingual Films
US8445526B2 (en) 2011-02-03 2013-05-21 Glaucoma & Nasal Therapies Llc Compositions and methods for treatment of glaucoma
EP2670368A4 (en) 2011-02-03 2015-04-15 Pharmedica Ltd New oral dissolving films for insulin administration, for treating diabetes
US20140163080A1 (en) 2011-02-03 2014-06-12 Gnt, Llc Compositions and Methods for Treatment of Glaucoma
US8703697B2 (en) 2011-06-24 2014-04-22 Luc QUINTIN Method for treating early severe diffuse acute respiratory distress syndrome
US8241661B1 (en) 2011-06-24 2012-08-14 Fuisz Richard C Biocompatible film with variable cross-sectional properties
US20130096170A1 (en) 2011-10-14 2013-04-18 Hospira, Inc. Methods of treating pediatric patients using dexmedetomidine
US20130116215A1 (en) 2011-10-28 2013-05-09 Mireia Coma Combination therapies for treating neurological disorders
EP3406247A1 (en) 2011-12-11 2018-11-28 Recro Pharma, Inc. Intranasal dexmedetomidine compositions and methods of use thereof
US8242158B1 (en) 2012-01-04 2012-08-14 Hospira, Inc. Dexmedetomidine premix formulation
CA2865593A1 (en) 2012-02-27 2013-09-06 Eye Therapies Llc Compositions and methods for the treatment of migraine
JP5921928B2 (en) 2012-03-28 2016-05-24 テルモ株式会社 Already diluted dexmedetomidine preparation
CN102657635B (en) 2012-05-04 2013-08-07 上海现代药物制剂工程研究中心有限公司 Spongy asenapine sublingual film agent with micropores and preparation method thereof
WO2013173317A1 (en) 2012-05-14 2013-11-21 Prospire, Llc TREATMENT OF OBSTRUCTIVE SLEEP APNEA WITH α2-ADRENERGIC RECEPTOR AGONISTS
EA201590165A1 (en) 2012-07-06 2015-08-31 Эгалет Лтд. CONTAINING ABUSE OF PHARMACEUTICAL COMPOSITIONS FOR CONTROLLED LIBERATION
US10130766B1 (en) 2012-08-08 2018-11-20 Neurowave Systems Inc. Intelligent pharmaceutical delivery system with automatic shutoff and method of using
SI2719376T1 (en) 2012-10-12 2015-06-30 Omya International Ag Gastroretentive drug formulation and delivery systems and their method of preparation using functionalized calcium carbonate
US20140180160A1 (en) 2012-10-12 2014-06-26 Emery N. Brown System and method for monitoring and controlling a state of a patient during and after administration of anesthetic compound
ES2659271T3 (en) * 2012-10-15 2018-03-14 Orion Corporation Veterinary method to relieve noise aversion
GB2510321B (en) 2012-11-12 2018-01-31 Biosurgical S L Agitation apparatus
WO2014116770A1 (en) 2013-01-23 2014-07-31 Arx, Llc Production of unit dose constructs
CN105228652A (en) * 2013-02-21 2016-01-06 罗契斯特大学 The garbage evaluating full cerebrovascular bypass footpath removes the method for function and the method based on this method treatment neurodegenerative disease
US20140261990A1 (en) 2013-03-15 2014-09-18 Monosol Rx, Llc Multi-layer films having uniform content
US9346601B2 (en) 2013-03-15 2016-05-24 Monosol Rx, Llc Reduction in stress cracking of films
US9303918B2 (en) 2013-03-15 2016-04-05 Monosol Rx, Llc Process for drying a wet film with control of loss on drying
WO2014144365A1 (en) 2013-03-15 2014-09-18 University Of Maryland, College Park Nano-liposomal formulations and methods of use
EP2976404B1 (en) 2013-03-22 2021-06-30 Adhesives Research, Inc. Hydrophilic adhesives and tapes and devices containing the same
EP2988666A1 (en) 2013-04-23 2016-03-02 The General Hospital Corporation System and method for monitoring anesthesia and sedation using measures of brain coherence and synchrony
WO2014176444A1 (en) 2013-04-24 2014-10-30 The General Hospital Corporation System and method for estimating high time-frequency resolution eeg spectrograms to monitor patient state
WO2014176441A1 (en) 2013-04-24 2014-10-30 The General Hospital Corporation System and method for monitoring level of dexmedatomidine-induced sedation
CN103284945A (en) 2013-06-03 2013-09-11 四川百利药业有限责任公司 Preparation method of pre-filled dexmedetomidine hydrochloride injection
JP6453319B2 (en) 2013-06-04 2019-01-16 モノソル リミテッド ライアビリティ カンパニー Water-soluble film seal solution, related methods, and related articles
US10314503B2 (en) 2013-06-27 2019-06-11 The General Hospital Corporation Systems and methods for tracking non-stationary spectral structure and dynamics in physiological data
US10383574B2 (en) 2013-06-28 2019-08-20 The General Hospital Corporation Systems and methods to infer brain state during burst suppression
US10602978B2 (en) 2013-09-13 2020-03-31 The General Hospital Corporation Systems and methods for improved brain monitoring during general anesthesia and sedation
JP6188933B2 (en) * 2013-10-07 2017-08-30 テイコク ファーマ ユーエスエー インコーポレーテッド Methods and compositions for transdermal delivery of non-sedating amounts of dexmedetomidine
EP3054934A4 (en) 2013-10-07 2017-04-05 Teikoku Pharma USA, Inc. Methods and compositions for treating withdrawal syndromes using non-sedative dexmedetomidine transdermal compositions
CN106456561B (en) 2013-10-07 2020-02-28 帝国制药美国公司 Methods and compositions for treating attention deficit hyperactivity disorder, anxiety disorders, and insomnia using dexmedetomidine transdermal compositions
EP3054931B1 (en) 2013-10-07 2020-12-23 Teikoku Pharma USA, Inc. Transdermal delivery device for managing pain comprising dexmedetomidine transdermal compositions
CA2924231C (en) 2013-10-07 2018-04-03 Teikoku Pharma Usa, Inc. Dexmedetomidine transdermal delivery devices and methods for using the same
WO2015069778A1 (en) 2013-11-05 2015-05-14 The General Hospital Corporation System and method for determining neural states from physiological measurements
US11033493B2 (en) 2013-12-02 2021-06-15 Intelgenx Corp. Film dosage form with extended release mucoadhesive particles
WO2015093503A1 (en) 2013-12-18 2015-06-25 丸石製薬株式会社 Water-containing adhesive patch
US20170274174A1 (en) 2014-08-22 2017-09-28 The General Hospital Corporation System and method for administering, monitoring and controlling biomimetic sleep
US20170273611A1 (en) 2014-08-22 2017-09-28 The General Hospital Corporation Systems and methods for discovery and characterization of neuroactive drugs
US20170231556A1 (en) 2014-08-22 2017-08-17 The General Hospital Corporation Systems and methods for predicting arousal to consciousness during general anesthesia and sedation
CN104161760A (en) 2014-09-01 2014-11-26 西北农林科技大学 Compound anesthetic for racoon dogs as well as preparation method and application thereof
MA41689A (en) 2014-10-15 2017-08-22 Bioxcel Corp PREVENTION OR TREATMENT OF SLEEP DISORDERS WITH A DEXMEDETOMIDINE FORMULATION
WO2016061554A1 (en) * 2014-10-16 2016-04-21 Bioxcel Corporation Synergistic composition of known, safe pharmaceuticals for use in insomnia and a method of treatment thereof
FI127534B (en) 2014-11-10 2018-08-31 Vetcare Oy COMPOSITIONS COMPRISING SUBSTITUTED BENZOFUROQUINOLIZINE AND ɑ2-ADRENERGIC AGONISTS
WO2016089997A1 (en) 2014-12-02 2016-06-09 Yale University Methods of preventing neurodegeneration of association cortex in a mammal
JP2016154598A (en) 2015-02-23 2016-09-01 ニプロ株式会社 Prefilled syringe filled with dexmedetomidine injection solution
CN104784174A (en) 2015-03-06 2015-07-22 北京大学第一医院 New pharmaceutical use of dexmedetomidine
US11160901B2 (en) 2015-04-10 2021-11-02 Tricol Biomedical, Inc. Bioadhesive chitosan gel for controlling bleeding and for promoting healing with scar reduction without obscuring or interfering with access to a surgical field
CN106038538A (en) 2015-04-17 2016-10-26 江苏恒瑞医药股份有限公司 Premixed preparation for dexmedetomidine
US10179136B2 (en) 2015-06-19 2019-01-15 Imprimis Pharmaceuticals, Inc. Pharmaceutical compositions and methods for anesthesiological applications
US20160374588A1 (en) 2015-06-24 2016-12-29 Microsoft Technology Licensing, Llc Monitoring hydration based on galvanic skin response
CN105168122B (en) 2015-09-24 2018-11-27 辰欣药业股份有限公司 A kind of dexmedetomidine hydrochloride injection and its preparation process
US20170128421A1 (en) 2015-11-11 2017-05-11 Siva Prasad Reddy Sura Premix formulation for parenteral use and packaging thereof
US10632043B2 (en) 2015-11-11 2020-04-28 Aurobindo Pharma Ltd Premix formulation for parenteral use and packaging thereof
EP3400050B1 (en) 2016-01-04 2020-12-23 Jurox Pty. Ltd. Drug release device and use
CN105534891A (en) 2016-01-18 2016-05-04 南京正科医药股份有限公司 Dexmedetomidine hydrochloride injection
US9949934B1 (en) 2016-10-20 2018-04-24 Intelgenx Corp. Device and method of treating conditions associated with neuroinflammation
US9717796B1 (en) 2016-04-20 2017-08-01 Slypharma, Llc Heat sterilizeable, premixed, ready to use dexmedetomidine solution packaged in a flexible plastic container
CN107412152B (en) 2016-05-24 2020-12-04 海南合瑞制药股份有限公司 Dexmedetomidine hydrochloride injection composition
WO2017213977A1 (en) 2016-06-07 2017-12-14 Mehra Akhil Methods and compositions for the treatment of trauma and stressor-related disorders
CN105997955B (en) 2016-06-28 2017-09-12 力品药业(厦门)有限公司 A kind of Palonosetron oral cavity membrane and preparation method thereof
RU2635532C1 (en) 2016-09-06 2017-11-13 Федеральное государственное бюджетное учреждение "Ростовский научно-исследовательский онкологический институт" Министерства здравоохранения Российской Федерации Method for prevention of agitation syndrome in children with oncological pathology
EP4241759A1 (en) 2016-10-12 2023-09-13 PS Therapy, Inc. Artificial tear, contact lens and drug vehicle compositions and methods of use thereof
JP7219712B2 (en) 2016-10-13 2023-02-08 キャタレント・ユーケー・スウィンドン・ザイディス・リミテッド Lyophilized pharmaceutical composition for vaginal delivery
JP6968184B2 (en) 2016-10-25 2021-11-17 キャタレント・ユーケー・スウィンドン・ザイディス・リミテッド Compositions of different densities for fast disintegrating multi-layer tablets
JP2019534288A (en) 2016-10-31 2019-11-28 テイコク ファーマ ユーエスエー インコーポレーテッド Method of pain management using dexmedetomidine transdermal delivery device
WO2018081792A2 (en) 2016-10-31 2018-05-03 Allodynic Therapeutics, Llc Combinations of opioid/tlr4 antagonists and acetaminophen for use in the treatment of emotional pain and insomnia
CN106727524A (en) 2016-11-08 2017-05-31 叶茂 A kind of pharmaceutical composition and its application
CN106727443A (en) 2016-12-08 2017-05-31 武汉大学 Preoperative calm stealthy nose paste preparation of a kind of pediatric anesthesia and preparation method thereof
CN106539778A (en) 2016-12-08 2017-03-29 武汉大学 Preoperative buccal oral cavity effervescent tablet of a kind of pediatric anesthesia and preparation method thereof
RU2759726C2 (en) 2016-12-13 2021-11-17 Орион Корпорейшн Dexmedetomidine or medetomidine for use in the treatment of separation anxiety in dogs
WO2018116202A1 (en) 2016-12-20 2018-06-28 Azista Industries Pvt Ltd Stabilized mouth freshener strips
KR20240031326A (en) * 2016-12-31 2024-03-07 바이오엑셀 테라퓨틱스 인코포레이티드 Use of Sublingual Dexmedetomidine for the treatment of Agitation
JP2020510675A (en) 2017-03-07 2020-04-09 チャイルズ, マークCHILDS, Marc Prevention of risks associated with drug-induced QT interval prolongation using specific inhibitors of the production of ROS of mitochondrial origin
CN107412204A (en) 2017-04-24 2017-12-01 重庆医科大学附属儿童医院 A kind of Foradil Aerolizer formoterol fumarate for being used to anaesthetize and preparation method thereof
CN107693485A (en) 2017-04-24 2018-02-16 重庆医科大学附属儿童医院 A kind of nasal drops for being used to anaesthetize and preparation method thereof
CN107137399A (en) 2017-04-24 2017-09-08 徐颖 A kind of sublingual tablet for being used to anaesthetize and preparation method thereof
CN107028880A (en) 2017-06-09 2017-08-11 安徽赛诺制药有限公司 A kind of production technology of dexmedetomidine hydrochloride parenteral solution
EP3668488A4 (en) 2017-08-15 2021-03-24 The Board of Trustees of the Leland Stanford Junior University Polymeric perfluorocarbon nanoemulsions for ultrasonic drug uncaging
CA3071552A1 (en) 2017-08-20 2019-02-28 Formulex Pharma Innovations Ltd. Dry powder compositions for intranasal delivery
US11857334B2 (en) 2017-10-04 2024-01-02 The General Hospital Corporation Systems and methods for monitoring a subject under the influence of drugs
US20190216345A1 (en) 2017-10-18 2019-07-18 Christoper Scheib Method and system for monitoring and displaying physiological conditions
US11147459B2 (en) 2018-01-05 2021-10-19 CareBand Inc. Wearable electronic device and system for tracking location and identifying changes in salient indicators of patient health
US11406316B2 (en) 2018-02-14 2022-08-09 Cerenion Oy Apparatus and method for electroencephalographic measurement
AU2019229784A1 (en) 2018-03-08 2020-08-13 Catalent U.K. Swindon Zydis Limited Process to reduce endotoxin in gelatin
KR20190109310A (en) 2018-03-15 2019-09-25 주식회사 바이오홀딩스 Nasal spray type formuation comprising levan for treating inflamentory diseases
CN108498469A (en) 2018-05-30 2018-09-07 宁波蒙曼生物科技有限公司 Dexmedetomidine hydrochloride freeze-dried powder and preparation method thereof
US20210267944A1 (en) 2018-06-27 2021-09-02 Bioxcel Therapeutics, Inc. Methods for treating agitation using dexmedetomidine hydrochloride
CA3103431A1 (en) 2018-06-27 2020-01-02 Bioxcel Therapeutics, Inc. Film formulations containing dexmedetomidine and methods of producing them
US11648197B2 (en) 2018-06-28 2023-05-16 Arx, Llc Dispensing method for producing dissolvable unit dose film constructs
CN110893186A (en) 2018-09-12 2020-03-20 宜昌人福药业有限责任公司 Pharmaceutical composition and preparation method and application thereof
JP2019048091A (en) 2018-10-23 2019-03-28 ニプロ株式会社 Prefilled syringe filled with dexmedetomidine injection solution
KR102183744B1 (en) 2018-11-26 2020-11-27 연세대학교 산학협력단 Methods for pedicting risk of delirium and devices for pedicting risk of delirium using the same
CN109620802A (en) 2018-12-05 2019-04-16 杜皓 A kind of anesthesia nasal spray and preparation method thereof
CN111481506B (en) 2019-01-25 2023-01-24 江苏恒瑞医药股份有限公司 Pharmaceutical product comprising a nasally administrable dexmedetomidine composition
US10900762B2 (en) 2019-02-21 2021-01-26 Aob Products Company Ammunition press and components thereof
CN112138250B (en) 2019-06-28 2023-04-14 四川普锐特药业有限公司 Medicament fluid dispenser and dexmedetomidine nasal spray device for maintaining uniform administration
CN112107544A (en) 2019-06-28 2020-12-22 四川普锐特药业有限公司 Dexmedetomidine nasal spray, preparation method and application thereof
CA3145388A1 (en) 2019-07-19 2021-01-28 Bioxcel Therapeutics, Inc. Non-sedating dexmedetomidine treatment regimens
CA3154659A1 (en) 2019-09-18 2021-03-25 Bioxcel Therapeutics, Inc. Systems and methods for detection and prevention of emergence of agitation
CA3167994A1 (en) 2020-02-14 2021-08-19 Bioxcel Therapeutics, Inc. Systems and methods for detection and prevention of emergence of agitation
EP4225305A1 (en) 2020-10-08 2023-08-16 Bioxcel Therapeutics, Inc. Treatment of bipolar disorders and psychosis using dexmedetomidine hydrochloride
EP4271376A1 (en) 2021-01-04 2023-11-08 BioXcel Therapeutics, Inc. Dexmedetomidine treatment regimens
EP4297871A1 (en) 2021-02-26 2024-01-03 BioXcel Therapeutics, Inc. Methods and compositions for treating agitation

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11786508B2 (en) 2016-12-31 2023-10-17 Bioxcel Therapeutics, Inc. Use of sublingual dexmedetomidine for the treatment of agitation
US11839604B2 (en) 2016-12-31 2023-12-12 Bioxcel Therapeutics, Inc. Use of sublingual dexmedetomidine for the treatment of agitation
US11931340B2 (en) 2016-12-31 2024-03-19 Bioxcel Therapeutics, Inc. Use of sublingual dexmedetomidine for the treatment of agitation
US11806429B2 (en) 2018-06-27 2023-11-07 Bioxcel Therapeutics, Inc. Film formulations containing dexmedetomidine and methods of producing them
US11890272B2 (en) 2019-07-19 2024-02-06 Bioxcel Therapeutics, Inc. Non-sedating dexmedetomidine treatment regimens

Also Published As

Publication number Publication date
JP2020503323A (en) 2020-01-30
TW202319048A (en) 2023-05-16
ZA202006636B (en) 2023-03-29
SG10202107367SA (en) 2021-08-30
MX2019007923A (en) 2019-09-10
AU2017388759B2 (en) 2021-11-11
EP3562486A1 (en) 2019-11-06
AU2017388759A1 (en) 2019-06-13
IL267689A (en) 2019-08-29
CN110337290A (en) 2019-10-15
US11931340B2 (en) 2024-03-19
JP2024014900A (en) 2024-02-01
TWI791479B (en) 2023-02-11
JP2021113203A (en) 2021-08-05
AU2022200892A1 (en) 2022-03-03
US20230218580A1 (en) 2023-07-13
JP6868698B2 (en) 2021-05-12
BR112019013503A2 (en) 2020-01-07
JP2022040229A (en) 2022-03-10
US11786508B2 (en) 2023-10-17
MX2022010274A (en) 2022-09-19
US20190365715A1 (en) 2019-12-05
WO2018126182A1 (en) 2018-07-05
US20230081503A1 (en) 2023-03-16
JP2021102654A (en) 2021-07-15
MX2023000213A (en) 2023-02-09
KR20190108104A (en) 2019-09-23
US20220031663A1 (en) 2022-02-03
JP6929479B2 (en) 2021-09-01
EP3562486A4 (en) 2020-08-19
KR20240031326A (en) 2024-03-07
CA3045043A1 (en) 2018-07-05
TW201829018A (en) 2018-08-16
US11839604B2 (en) 2023-12-12
EP3562486B1 (en) 2024-03-13
JP7008153B2 (en) 2022-02-10

Similar Documents

Publication Publication Date Title
US11786508B2 (en) Use of sublingual dexmedetomidine for the treatment of agitation
EP2429521B1 (en) Sublingual dexmedetomidine compositions and methods of use thereof
KR101077468B1 (en) Stable orodispersible film formulation
US20230414572A1 (en) Prevention or treatment of sleep disorders using dexmedetomidine formulation
EP4076380B1 (en) Transmucosal therapeutic system containing agomelatine
WO2022183029A1 (en) Methods and compositions for treating agitation
TWI833550B (en) Use of sublingual dexmedetomidine for the treatment of agitation
US20050089558A1 (en) Compositions and methods for the co-formulation and administration of tramadol and propoxyphene
EA042166B1 (en) USE OF SUBLINGUAL DEXMEDETOMIDINE FOR THE TREATMENT OF AGITATION
NZ794716A (en) Use of sublingual Dexmedetomidine for the treatment of agitation
NZ794535A (en) Use of sublingual Dexmedetomidine for the treatment of agitation
NZ753804A (en) Use of sublingual dexmedetomidine for the treatment of agitation
US20240139169A1 (en) Methods and compositions for treating agitation
EP3915548A1 (en) Transmucosal pharmaceutical compositions of antiviral drugs

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: SENT TO CLASSIFICATION CONTRACTOR

AS Assignment

Owner name: E. Z. BIOXCEL SOLUTIONS PVT. LTD., INDIA

Free format text: INVENTION ASSIGNMENT AGREEMENT (EMPLOYMENT);ASSIGNOR:SAINI, DEEPA;REEL/FRAME:064441/0702

Effective date: 20140916

Owner name: E. Z. BIOXCEL SOLUTIONS PVT. LTD., INDIA

Free format text: INVENTION ASSIGNMENT AGREEMENT (EMPLOYMENT);ASSIGNOR:NEGI, HARSH;REEL/FRAME:063040/0828

Effective date: 20140916

Owner name: BIOXCEL THERAPEUTICS, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:E. Z. BIOXCEL SOLUTIONS PVT. LTD.;REEL/FRAME:062952/0205

Effective date: 20220419

Owner name: E. Z. BIOXCEL SOLUTIONS PVT. LTD., INDIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SHARMA, SAMEER;REEL/FRAME:062952/0198

Effective date: 20190903

Owner name: BIOXCEL THERAPEUTICS, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:YOCCA, FRANK;REEL/FRAME:062952/0170

Effective date: 20191015

Owner name: BIOXCEL THERAPEUTICS, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BIOXCEL CORPORATION;REEL/FRAME:062952/0147

Effective date: 20190904

Owner name: BIOXCEL CORPORATION, CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NANDABALAN, KRISHNAN;REEL/FRAME:062952/0101

Effective date: 20190904

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

Free format text: SPECIAL NEW

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

Free format text: NON FINAL ACTION MAILED

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

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