WO2024097387A1 - Compositions pour administration intraveineuse - Google Patents

Compositions pour administration intraveineuse Download PDF

Info

Publication number
WO2024097387A1
WO2024097387A1 PCT/US2023/036747 US2023036747W WO2024097387A1 WO 2024097387 A1 WO2024097387 A1 WO 2024097387A1 US 2023036747 W US2023036747 W US 2023036747W WO 2024097387 A1 WO2024097387 A1 WO 2024097387A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
compound
disease
pain
condition
Prior art date
Application number
PCT/US2023/036747
Other languages
English (en)
Inventor
Russell Birch Poe
Original Assignee
Astrocyte Pharmaceuticals, 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 Astrocyte Pharmaceuticals, Inc. filed Critical Astrocyte Pharmaceuticals, Inc.
Publication of WO2024097387A1 publication Critical patent/WO2024097387A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to compounds, compositions, and methods of use thereof for treating, ameliorating, or promoting recovery from certain conditions of the brain, central nervous system (CNS), or cardiovascular system such as a brain injury, a neurodegenerative condition, or cardiac ischemia.
  • CNS central nervous system
  • cardiovascular system such as a brain injury, a neurodegenerative condition, or cardiac ischemia.
  • BACKGROUND OF THE INVENTION [0004] Brain injuries are a distressingly common medical condition and one of the leading causes of morbidity and mortality worldwide. The brain is particularly susceptible to injury as neurons have a limited capacity to repair. When an individual is born, the brain already has essentially all the neurons it will have in life. Unlike other cells in the body, neurons stop reproducing shortly after birth.
  • Heart disease and stroke statistics-2015 update a report from the American Heart Association,” Circulation.2015 ;e29-322).
  • the site of insult in the brain typically contains a core of tissue that is irreversibly damaged, and then also an area of viable but at-risk tissue called the penumbra.
  • the penumbra an area of viable but at-risk tissue.
  • the insufficient oxygen and glucose supply to brain cells results in further secondary injury to the penumbra.
  • the lack of oxygen and glucose decreases energy production by cell mitochondria.
  • astrocyte function plays a key role in broader neurorestoration – in the period 24-96 hours following brain insult, in the period months-years in patients with neurodegeneration such as Alzheimer’s, or most generally in aged individuals.
  • the inability of brain cells to regenerate requires the remaining intact brain tissue to reorganize in an attempt to recover any loss of function. This potential for neural reorganization is diminished in older individuals.
  • GPCR receptors have been suggested to mediate cardioprotective effects. Therefore, there is potential to treat heart and cardiovascular conditions by similar mechanisms of action via modulation of these receptors.
  • the present disclosure provides an injectable liquid pharmaceutical composition, comprising: (a) compound A: at a concentration of about 5 mg/mL to about 75 mg/mL; (b) 30-60% v/v glycol solvent in water; and (c) a buffer. [0011] Various embodiments and additional aspects of the present disclosure are described below.
  • the present invention provides pharmaceutical compositions for injectable, e.g., intravenous, administration.
  • injectable e.g., intravenous
  • many active pharmaceutical agents are not orally bioavailable.
  • the amount of the API that is ultimately absorbed and distributed to the site(s) of action in the body is greatly influenced by the pharmaceutical formulation used.
  • Discovering an optimal injectable formulation presents various challenges including solubilizing and stabilizing the API in a sufficiently small volume so that injection is possible.
  • a formulation that has been successful for one API will not necessarily be successful for a different API, even a structurally- similar one.
  • the present invention provides liquid oral pharmaceutical compositions comprising an agonist of the adenosine receptor 1 (A1R) and/or adenosine receptor 3 (A3R).
  • A1R adenosine receptor 1
  • A3R adenosine receptor 3
  • United States Patent Nos. 9,789,131 the entirety of which is hereby incorporated herein by reference, describe certain therapeutically beneficial compounds. Such compounds include compound A: 3 BUSINESS.30655163.1 393522-004WO (204863) A.
  • Compound A is designated as MRS4322 in the ’131 patent and the synthesis of compound A is described in detail at Example 9 of the ’131 patent, which is hereby incorporated by reference in its entirety.
  • Compound A may also be prepared by the convergent synthesis described in PCT/US2022/071971, which is hereby incorporated by reference.
  • Compound A has shown effectiveness in treatment of diseases such as TBI and stroke. See, e.g.: 1. Liston TE, Hama A, Boltze J, et al. Adenosine A1R/A3R (Adenosine A1 and A3 Receptor) Agonist AST-004 Reduces Brain Infarction in a Nonhuman Primate Model of Stroke. Stroke. 2022;53(1):238-248. Epub 2021/11/23. doi: 10.1161/strokeaha.121.036396. PubMed PMID: 34802248. 2.
  • Form A or Form B solid form of compound A is used in the manufacture of a disclosed pharmaceutical composition.
  • Form A and Form B are described in PCT Publication WO 2020/069068, which is hereby incorporated by reference.
  • the present invention provides compositions for intravenous (IV) administration of compound A.
  • the composition is administered via a subcutaneous (SC/SQ), intraperitoneal (IP), intravenous (IV), intradermal (ID), or intramuscular (IM) route.
  • SC/SQ subcutaneous
  • IP intraperitoneal
  • IV intravenous
  • ID intradermal
  • IM intramuscular
  • the composition is administered parenterally.
  • the compound or pharmaceutically acceptable salt thereof is administered intravenously.
  • the compound or pharmaceutically acceptable salt thereof is administered as a continuous intravenous (IV) infusion.
  • the compound or pharmaceutically acceptable salt thereof is administered initially as an IV bolus, followed by continuous IV infusion, e.g., to maintain a desired plasma concentration.
  • the compound or pharmaceutically acceptable salt thereof is administered as a slow bolus/rapid infusion.
  • the slow bolus/rapid infusion comprises IV administration over an about 5-60 minute period, e.g., a 5-30, 5-20, 10-20, or about 10 minute period.
  • the slow bolus/rapid infusion is then followed by continuous IV infusion.
  • the slow bolus/rapid infusion followed by continuous IV infusion is for treating TBI or stroke in a subject.
  • the slow bolus/rapid infusion (whether or not followed by a continuous IV infusion) consists of about 5 mg to about 500 mg of Compound A.
  • the slow bolus/rapid infusion consists of about 15 mg to about 375 mg of Compound A.
  • the slow bolus/rapid infusion consists of about 25 mg to about 125 mg of Compound A.
  • the slow bolus/rapid infusion consists of about 75 mg to about 180 mg of Compound A. In certain embodiments, the slow bolus/rapid infusion consists of about 100 mg to about 150 mg of Compound A. In some embodiments, the slow bolus/rapid infusion consists of about 5 mg to about 130 mg of Compound A. [0020] In certain embodiments, the slow bolus/rapid infusion consists of about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, or about 200 mg of Compound A. In certain embodiments, the slow bolus/rapid infusion consists of about 150 mg of Compound A.
  • the compound or composition is administered over a 6-hour period. In some embodiments, the compound or composition is administered over an 8-hour period. In some embodiments, the compound or composition is administered over a 4-hour period. In some embodiments, the compound or composition is administered over a 2-hour period. In some embodiments, the compound or composition is administered over a 1-hour period. [0022] In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 1000 ng/mL and 6000 ng/mL.
  • the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 1000 ng/mL and 5000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 1000 ng/mL and 4000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 1000 ng/mL and 3000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 1000 ng/mL and 2000 ng/mL.
  • the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 2000 ng/mL and 6000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 3000 ng/mL and 6000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 4000 ng/mL and 6000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 5000 ng/mL and 6000 ng/mL.
  • the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 2000 ng/mL and 6000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 3000 ng/mL and 6000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 4000 ng/mL and 6000 ng/mL. In some embodiments, the compound or composition is administered at a dose, wherein the dose achieves a plasma concentration level of between 5000 ng/mL and 6000 ng/mL.
  • the method comprises the steps: A) administering a single bolus comprising Compound A; and B) administering an infusion of a pharmaceutical 6 BUSINESS.30655163.1 393522-004WO (204863) composition comprising Compound A over a period of 1 to 6 hours.
  • the method comprises the steps: A) administering a single bolus comprising Compound A; and B) administering an infusion of a pharmaceutical composition comprising Compound A over a period of 1 to 5 hours.
  • the method comprises the steps: A) administering a single bolus comprising Compound A; and B) administering an infusion of a pharmaceutical composition comprising Compound A over a period of 1 to 4 hours.
  • the method comprises the steps: A) administering a single bolus comprising Compound A; and B) administering an infusion of a pharmaceutical composition comprising Compound A over a period of 1 to 3 hours. In certain embodiments, the method comprises the steps: A) administering a single bolus comprising Compound A; and B) administering an infusion of a pharmaceutical composition comprising Compound A over a period of 1 to 2 hours.
  • the single bolus comprises about 5 mg to about 500 mg of Compound A. In certain embodiments, the single bolus comprises about 15 mg to about 375 mg of Compound A. In certain embodiments, the single bolus comprises about 25 mg to about 125 mg of Compound A.
  • the single bolus comprises about 75 mg to about 180 mg of Compound A. In certain embodiments, the single bolus comprises about 100 mg to about 150 mg of Compound A. In some embodiments, the single bolus comprises about 5 mg to about 130 mg of Compound A. [0027] In certain embodiments, the single bolus comprises about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, or about 200 mg of Compound A. In certain embodiments, the single bolus comprises about 150 mg of Compound A. [0028] In certain embodiments, the infusion comprises a dose of about 600 mg to about 2900 mg of Compound A.
  • the infusion comprises a dose of about 700 mg to about 2800 mg of Compound A. In certain embodiments, the infusion comprises a dose of about 800 mg to about 2700 mg of Compound A. In certain embodiments, the infusion comprises a dose of about 900 mg to about 2600 mg of Compound A. In certain embodiments, the infusion comprises a dose of about 1000 mg to about 2500 mg of Compound A. In certain embodiments, the infusion comprises a dose of about 1100 mg to about 2400 mg of Compound A. In certain embodiments, the infusion comprises a dose of about 500 mg to about 1800 mg of Compound A. In certain embodiments, the infusion comprises a dose of about 600 mg to about 1700 mg of Compound A.
  • the infusion comprises a dose of about 700 mg to about 1600 mg of Compound A.
  • the infusion 7 BUSINESS.30655163.1 393522-004WO (204863) comprises a dose of about 800 mg to about 1500 mg of Compound A.
  • the infusion comprises a dose of about 900 mg to about 1400 mg of Compound A.
  • the infusion comprises a dose of about 1000 mg to about 1300 mg of Compound A.
  • the infusion comprises a dose of about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg, about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg of Compound A.
  • the total dose of Compound A administered is about 750 mg to about 3000 mg.
  • the total dose of Compound A administered is about 1000 mg to about 2500 mg. In certain embodiments, the total dose of Compound A administered is about 1250 mg to about 2000 mg. In certain embodiments, the total dose of Compound A administered is about 1300 mg to about 1800 mg. In certain embodiments, the total dose of Compound A administered is about 1000 mg to about 1500 mg.
  • the total dose of Compound A administered is about 500 mg, about 600 mg, about 700 mg, about 750 mg, about 800 mg, about 900 mg, about 1000 mg, about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg of Compound A.
  • Compound A is administered at a higher (loading) dose sufficient to achieve a desired plasma concentration that is then followed by a continuous infusion (maintenance dose) over a period of time such as about 6 hours or 1-6 hours.
  • the loading dose is administered using an IV bag at a higher concentration and the maintenance dose is administered using a second IV bag in which Compound A is at a lower concentration.
  • the loading dose is administered using a faster rate of infusion and the maintenance dose is administered using the same IV bag at a slower rate of infusion.
  • the present disclosure provides an injectable liquid pharmaceutical composition, comprising: 8 BUSINESS.30655163.1 393522-004WO (204863) (a) compound A: at a concentration of about 5 (b) 30-60% v/v glycol solvent in water; and (c) a buffer. [0035] In some embodiments, the concentration of compound A is about 15 mg/mL to about 50 mg/mL. In some embodiments, the concentration of compound A is about 25 mg/mL. [0036] In some embodiments, the composition comprises 30-50% v/v glycol solvent in water. [0037] In some embodiments, the composition does not comprise a glycol solvent.
  • the concentration of compound A is about 15 mg/mL to about 50 mg/mL, about 16 mg/mL to about 45 mg/mL, about 17 mg/mL to about 40 mg/mL, about 16 mg/mL to about 35 mg/mL, about 15 mg/mL to about 35 mg/mL, or about 20 mg/mL to about 30 mg/mL.
  • the concentration of compound A is about 15 mg/mL, about 20 mg/mL, about 21 mg/mL, about 22 mg/mL, about 23 mg/mL, about 24 mg/mL, about 25 mg/mL, about 26 mg/mL, about 27 mg/mL, about 28 mg/mL, about 29 mg/mL or about 30 mg/mL. In some embodiments, the concentration of compound A is about 25 mg/mL.
  • a glycol solvent is any solvent comprising repeating polyether units, e.g., - ((CH 2 ) n O)-, where n is an integer from 2 to 50,000.
  • Glycol ethers are either “e-series” or “p- series” glycol ethers, depending on whether they are made from ethylene oxide or propylene oxide, respectively.
  • e-series glycol ethers are found in pharmaceuticals, sunscreens, cosmetics, inks, dyes and water-based paints, while p-series glycol ethers are used in degreasers, cleaners, aerosol paints and adhesives.
  • Both E-series glycol ethers and P-series glycol ethers can be used as intermediates that undergo further chemical reactions, producing glycol diethers and glycol ether acetates.
  • P-series glycol ethers are marketed as having lower toxicity than the E-series.
  • the glycol solvent comprises a dialkylene glycol monoalkyl ether, such as, e.g., diethylene glycol monoethyl ether.
  • a solvent is an alkylene glycol solvent.
  • the alkylene glycol solvent is ethylene glycol, propylene glycol, butylene glycol, or the like.
  • the glycol solvent is propylene glycol.
  • a solvent is a glycol ether.
  • a solvent is a dialkylene glycol monoalkyl ether, such as, e.g., diethylene glycol monoethyl ether.
  • a dialkylene glycol monoalkyl ether such as, e.g., diethylene glycol monoethyl ether.
  • glycol solvent comprises a polyethylene glycol.
  • the glycol solvent comprises propylene glycol.
  • the glycol solvent is ethylene glycol monomethyl ether (2- methoxyethanol, CH3OCH2CH2OH), ethylene glycol monoethyl ether (2-ethoxyethanol, CH3CH2OCH2CH2OH), ethylene glycol monopropyl ether (2-propoxyethanol, CH3CH2CH2OCH2CH2OH), ethylene glycol monoisopropyl ether (2-isopropoxyethanol, (CH3)2CHOCH2CH2OH), ethylene glycol monobutyl ether (2-butoxyethanol, CH 3 CH 2 CH 2 CH 2 OCH 2 CH 2 OH), ethylene glycol monophenyl ether (2-phenoxyethanol, C6H5OCH2CH2OH), ethylene glycol monobenzyl ether (2-benzyloxyethanol, C 6 H 5 CH 2 OCH 2 CH 2 OH), propylene glycol methyl ether, (1-methoxy-2-propanol, CH3OCH2CH(OH)CH3), diethylene glycol methyl ether, (1-
  • the polyethylene glycol is selected from PEG 200, PEG 300, PEG 400, SR PEG 400, PEG 540, PEG 600, PEG 900, or a mixture of any of the foregoing.
  • the polyethylene glycol is PEG 400.
  • a composition of the present invention comprises more than one solvent, wherein at least one of the more than one solvents is an alcohol solvent and one of the more than one solvents is a glycol solvent.
  • an alcohol solvent and a glycol solvent are each present in an amount of about 15 wt% to about 35 wt%.
  • an alcohol solvent and a glycol solvent are each present in an amount of about 15 wt% to about 30 wt%. In certain embodiments, an alcohol solvent and a glycol solvent are each present in an amount of about 15 wt% to about 25 wt%. In certain embodiments, an alcohol solvent and a glycol solvent are each present in an amount of about 18 wt% to about 21 wt%.
  • the alcohol solvent is an alkyl alcohol (e.g., ethanol) present in an amount of about 1-25 wt% and the glycol solvent is an alkylene glycol (e.g., propylene glycol) present in an amount of about 5-35 wt%.
  • the alcohol solvent is ethanol and is present in an amount of about 1-10 wt% and the glycol solvent is propylene glycol and is present in an amount of about 10-30 wt%.
  • Glycol solvents are vulnerable to oxidation and can generate peroxides, such as H2O2, resulting in discoloration of the IV solution.
  • the polyethylene glycol is substantially free of peroxides and other degradants.
  • the polyethylene glycol contains ⁇ 0.04% peroxides, ⁇ 0.03% peroxides, ⁇ 0.02% peroxides, or ⁇ 0.01% peroxides.
  • the polyethylene glycol contains about 1.0 meqO2/kg (milliequivalents/kg) or less of peroxide content.
  • the polyethylene glycol contains about 0.9, 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, or 0.1 meqO2/kg or less of peroxide content. In some embodiments, the polyethylene glycol contains 100 ppm meqO 2 /kg or less of peroxide content.
  • the glycol solvent comprises 35-50% v/v of a polyethylene glycol. In some embodiments, the glycol solvent comprises 35-45% v/v, or 35-40% v/v of a polyethylene glycol. [0053] In some embodiments, the glycol solvent comprises about 40% v/v of a polyethylene glycol.
  • the glycol solvent comprises about 30% v/v, 35% v/v, 45% v/v ⁇ 50% v/v ⁇ 55% v/v ⁇ or 65% v/v of a polyethylene glycol.
  • the polyethylene glycol has an average molecular weight (MW) of 200-600. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 200-600. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 250-350. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 350-450.
  • the polyethylene glycol has an average molecular weight (MW) of 450-550. In some embodiments, the polyethylene 11 BUSINESS.30655163.1 393522-004WO (204863) glycol has an average molecular weight (MW) of 550-650. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 650-750. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 750-850. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 850-950. In some embodiments, the polyethylene glycol has an average molecular weight (MW) of 380- 420.
  • a solvent other than a glycol solvent is used. Any pharmacueutically acceptable solvent that enhances the solubility of Compound A in an aqueous solution may be used.
  • the solvent is selected from a cyclodextrin, a mannitol solution, benzylbenzoate, an alcohol such as ethanol or benzyl alcohol, or n-methyl pyrrolidine (NMP).
  • NMP n-methyl pyrrolidine
  • the buffer is a citrate, phosphate, borate, or acetate buffer.
  • the buffer is a phosphate buffer.
  • the composition has a pH of about 6.0 to about 9.0. the composition has a pH of about 7.0 to about 8.5. In some embodiments, the composition has a pH of about 7.5 to about 8.3.
  • the composition has a pH of about 7.8.
  • the composition further comprises an antioxidant.
  • the antioxidant is sodium metabisulfite.
  • the antioxidant is selected from BHT and BHA.
  • the composition after storage at 25 °C and 60% relative humidity (RH) for a period of 6 months, the composition contains an amount of compound A within 0.5% of the amount of compound A of initial manufacture of the composition. In some embodiments, the composition contains less than 0.1% impurities after storage at 25 °C and 60% RH for a period of 6 months.
  • the pH of the composition after storage at 25 °C and 60% RH for a period of 6 months remains within 0.2 units of the pH measured at initial manufacture of the composition. 12 BUSINESS.30655163.1 393522-004WO (204863) [0064] In some embodiments, after storage at 40 °C and 75% relative humidity (RH) for a period of 1 month, the composition contains an amount of compound A within 0.5% of the amount of compound A of initial manufacture of the composition. [0065] In some embodiments, the composition contains less than 0.1% impurities after storage at 40 °C and 75% RH for a period of 1 month.
  • the pH of the composition after storage at 40 °C and 75% RH for a period of 1 month remains within 0.2 units of the pH measured at initial manufacture of the composition.
  • impurities from manufacture of Compound A may oxidize.
  • the composition contains no more than 0.8% by HPLC, as compared to the AUC (area under the curve) of compound A, of the following compound:
  • an injectable liquid pharmaceutical composition comprising: (a) compound A: at a concentration of about 5 mg/mL to about 75 mg/mL; (b) a buffer; and (c) optionally, a pharmaceutically acceptable solvent.
  • the present disclosure provides a unit dosage form comprising a pharmaceutical composition disclosed herein.
  • the unit dosage form contains about 15 mg to about 500 mg of compound A.
  • the unit dosage form contains about 25 mg to about 375 mg of compound A.
  • the unit dosage form contains about 25 mg, about 125, or about 375 mg of compound A.
  • the unit dosage form contains about 125 mg of compound A in a fill volume of about 5 mL.
  • the unit dosage form is packaged in a glass or plastic vial. In some embodiments, the unit dosage form is packaged in an IV bag.
  • the unit dosage form is packaged in a pre-filled syringe. In some embodiments, the unit dosage form is packaged as a dry powder for reconstitution.
  • the present invention provides a method of treating an injury, disease, or condition selected from traumatic brain injury (TBI), concussion, stroke, polytrauma, cardiac arrest, near drowning, altitude sickness, brain injuries from directed energy or Havanna Syndrome, partial or total spinal cord transection, malnutrition, toxic neuropathies, meningoencephalopathies, neurodegeneration caused by a genetic disorder, age-related neurodegeneration, vascular disease, Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Huntington’s Disease (HD), Multiple Sclerosis (MS), amyotrophic lateral sclerosis (ALS), chronic traumatic encephalopathy (CTE), cardiovascular disease, autoimmune diseases, allergic diseases, transplant rejection, graft-versus-host disease, intraocular hypertension, glaucoma, odor sensitivity, an olfactor
  • the injury, disease, or condition is selected from acute pain, chronic pain, nociceptive pain, neuropathic pain, inflammatory pain, and acute pain.
  • the pain is selected from musculoskeletal pain, fibromyalgia, myofascial pain, pain during menstruation, pain during osteoarthritis, pain during rheumatoid arthritis, pain during gastrointestinal inflammation, pain during inflammation of the heart muscle, pain during multiple sclerosis, pain during neuritis, pain during AIDS, pain during 14 BUSINESS.30655163.1 393522-004WO (204863) chemotherapy, tumor pain, headache, chronic pain syndrome (CPS), central pain, trigeminal neuralgia, shingles, stamp pain, phantom limb pain, temporomandibular joint disorder, nerve injury, migraine, post-herpetic neuralgia, neuropathic pain encountered as a consequence of injuries, amputation infections, metabolic disorders or degenerative diseases of the nervous system, n
  • the injury, disease, or condition is selected from traumatic brain injury (TBI), stroke, a neurodegenerative condition, and a heart and cardiovascular disease.
  • TBI traumatic brain injury
  • the injury, disease, or condition is TBI selected from concussion, blast injury, combat-related injury, a mild, moderate or severe blow to the head, whiplash, sports-related injury, or a head injury sustained from a fall or other accident.
  • the injury, disease, or condition is a stroke selected from ischemic stroke, hemorrhagic stroke, subarachnoid hemorrhage, cerebral vasospasm, and transient ischemic attacks (TIA).
  • the compound or composition is administered within 24 hours of the TBI or stroke. [0078] In some embodiments, the compound or composition is administered within 8 hours of the TBI or stroke. [0079] In some embodiments, the compound or composition is administered at least during the first 8-48 hours following the TBI or stroke. [0080] In some embodiments, neuroprotection or neurorestoration is increased in the patient as compared with an untreated patient.
  • the injury, disease, or condition is a neurodegenerative disease selected from Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Huntington’s Disease (HD), Multiple Sclerosis (MS), amyotrophic lateral sclerosis (ALS), chronic traumatic encephalopathy (CTE), and a neurodegenerative condition caused by a virus, alcoholism, tumor, toxin, or repetitive brain injuries.
  • AD Alzheimer’s Disease
  • PD Parkinson’s Disease
  • HD Huntington’s Disease
  • MS Multiple Sclerosis
  • ALS amyotrophic lateral sclerosis
  • CTE chronic traumatic encephalopathy
  • a neurodegenerative condition caused by a virus, alcoholism, tumor, toxin, or repetitive brain injuries.
  • the injury, disease, or condition is heart or cardiovascular disease selected from cardiac ischemia, myocardial infarction, a cardiomyopathy, coronary artery disease, arrhythmia, myocarditis, pericarditis, angina, hypertensive heart disease, endocarditis, rheumatic heart disease, congenital heart disease, and atherosclerosis.
  • cardiovascular disease selected from cardiac ischemia, myocardial infarction, a cardiomyopathy, coronary artery disease, arrhythmia, myocarditis, pericarditis, angina, hypertensive heart disease, endocarditis, rheumatic heart disease, congenital heart disease, and atherosclerosis.
  • the present invention provides a method of increasing neuroprotection or neurorestoration in a patient in need thereof who has suffered a TBI or stroke, comprising administering to the patient an effective amount of a composition described herein.
  • the present invention provides a method of treating an injury, disease, disorder, or condition selected from: (i) brain damage caused by radiation or collateral brain damage associated with radiation cancer therapy or migraine treatment; (ii) migraine headache; (iii) a condition associated with a brain injury or a neurodegenerative condition; and (iv) an autoimmune disease or condition, glaucoma, an otic disorder, progressive hearing loss, tinnitus, epilepsy, pain control, pain mediated by the CNS, neuropathic pain, inflammatory pain, or acute pain; comprising administering to a patient in need thereof an effective amount of a composition described herein.
  • an injury, disease, disorder, or condition selected from: (i) brain damage caused by radiation or collateral brain damage associated with radiation cancer therapy or migraine treatment; (ii) migraine headache; (iii) a condition associated with a brain injury or a neurodegenerative condition; and (iv) an autoimmune disease or condition, glaucoma, an otic disorder, progressive hearing loss, tinnitus,
  • the composition increases neuroprotection or neurorestoration in the patient as compared with an untreated patient.
  • the condition associated with a brain injury or a neurodegenerative condition is selected from epilepsy, migraine, collateral brain damage associated with radiation cancer therapy, depression, mood or behavioral changes, dementia, erratic behavior, suicidality, tremors, Huntington’s chorea, loss of coordination of movement, deafness, impaired speech, dry eyes, hypomimia, attention deficit, memory loss, cognitive difficulties or deficit in cognition, deficit in CNS function, deficit in learning, vertigo, dysarthria, dysphagia, ocular abnormalities, and disorientation.
  • the injury, disease, or condition is migraine.
  • the injury, disease, or condition is pain selected from central pain syndrome, peripheral neuropathy, corneal neuropathic pain, post stroke pain, and pain caused by multiple sclerosis.
  • the present invention provides a method of increasing cardioprotection or regeneration of damaged heart tissue in a patient in need thereof who has suffered a cardiac ischemia or myocardial infarction, comprising administering to the patient an effective amount of a composition described herein.
  • the present invention provides a method of treating an addiction, addictive behavior, behavioral addiction, brain reward system disorder, or a 16 BUSINESS.30655163.1 393522-004WO (204863) compulsive disorder, comprising administering to a patient in need thereof an effective amount of a composition described herein.
  • the present invention provides a method of treating a disease, disorder, or condition selected from deficit in cognition, deficit in CNS function, deficit in learning, and memory loss, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable composition thereof.
  • the disease, disorder, or condition is deficit in cognition.
  • the disease, disorder, or condition is deficit in CNS function.
  • the disease, disorder, or condition is deficit in learning.
  • the disease, disorder, or condition is memory loss.
  • the subject has suffered one or more traumatic brain injuries (TBI or TBIs) and the disease, disorder, or condition is associated with the TBI or TBIs.
  • TBI or TBIs traumatic brain injuries
  • the subject has suffered one or more strokes and the disease, disorder, or condition is associated with the one or more strokes.
  • the subject has suffered one or more ischemic strokes, hemorrhagic strokes, subarachnoid hemorrhages, cerebral vasospasms, or transient ischemic attacks (TIA).
  • TIA transient ischemic attacks
  • the subject has Alzheimer’s disease and the disease, disorder, or condition is associated with the Alzheimer’s disease.
  • a method provided herein improves cognitive or neurological function as measured by a score increase between about 1% and 40% in the delayed verbal recall task of the revised Wechsler Memory Scale.
  • a method provided herein improves the score between about 5-10%, 10-20%, 15-30%, 20-30%, 30-40%, or 5-30% in the delayed verbal recall task of the revised Wechsler Memory Scale.
  • the method increases synaptic plasticity, improves hippocampal long-term potentiation, improves cognitive function, decreases cognitive impairment, and/or improves or restores memory or learning.
  • the method increases synaptic plasticity, improves hippocampal long-term potentiation, improves cognitive function, decreases cognitive impairment, prevents or delays cognitive decline, decreases plaque burden, enhances beta amyloid clearance, and/or improves or restores memory or learning.
  • the method improves or enhances cognition or neurological function by enhancing synaptogenesis.
  • the present invention provides a method of improving cognitive or neurological function in a subject having Alzheimer’s disease, comprising administering to a subject in need thereof an effective amount of Compound A, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable composition thereof.
  • the improvement in cognitive or neurological function as measured by a score increase between about 1% and 40%, or about 5-10%, 10-20%, 15-30%, 20-30%, 30-40%, or 5-30%, in the delayed verbal recall task of the revised Wechsler Memory Scale.
  • treatment refers to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein.
  • treatment is administered after one or more symptoms have developed.
  • treatment is administered in the absence of symptoms.
  • treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors).
  • the present invention provides a method of preventing and/or treating brain damage associated with acute brain trauma as well as longer term diseases of the brain and CNS and heart and cardiovascular diseases and conditions.
  • the present invention provides methods of treating such injuries, diseases, and conditions by utilizing neuroprotective and neurorestorative effects mediated by astrocytes, which are now understood as the key natural caretaker cell of neurons, as well as the astrocyte mitochondria, which supply a significant portion of the brain’s energy.
  • the present invention provides methods of treating such injuries, diseases, and conditions by cardioprotective and regenerative effects mediated by A3R receptors.
  • astrocyte caretaker functions such as their neuroprotective and neurorestorative functions, in turn enhancing the resistance of neurons and other cells to both acute injury and long-term stress.
  • the present invention provides compounds and methods of use thereof for treating, ameliorating, or promoting recovery from certain conditions of the brain or central nervous system (CNS) such as brain injuries, for example by increasing neuroprotection and/or neurorestorative effects mediated by astrocytes, glia, microglia, neurons, endothelium cells or other cells of the brain and/or CNS, comprising administering to a patient in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • CNS central nervous system
  • Astrocytes play key roles in supporting and protecting neurons and they critically affect the outcome of brain injuries that cause brain damage, such as ischemic injuries.
  • the central role astrocyte mitochondria themselves play in these brain functions is less well appreciated. For example, inhibition of astrocyte mitochondria increases swelling and leads to necrotic cell death. Neurons are permanently injured by recurrent spreading depolarizations only if astrocyte mitochondrial function fails, and astrocyte mitochondria are required for reduction of pathophysiological elevations of extracellular K + , which initiate spreading depolarizations.
  • Activation of purinergic receptors on astrocytes results in increased mitochondrial Ca 2+ that enhances mitochondrial citric acid cycle function and increases respiration and ATP production.
  • the present invention relates to the discovery that activation of astrocyte purinergic receptors enhances brain cell survival signalling pathways, enabling both astrocyte and neuronal viability during oxidative stress. Furthermore, activated astrocytes generate and supply reduced glutathione, a key antioxidant that aids in the resistance of both astrocytes and neurons to oxidative stress.
  • the present invention provides a method of modulating astrocyte purinergic receptors to promote survival and viability of one or more cell types in the brain of a patient after oxidative stress, such as oxidative stress caused by a brain injury, ischemia-reperfusion or a neurodegenerative condition, comprising administering to a patient in need thereof a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • oxidative stress such as oxidative stress caused by a brain injury, ischemia-reperfusion or a neurodegenerative condition
  • activation of astrocytes is achieved through contacting with a disclosed compound one or more purinergic receptors such as adenosine receptors (ARs), for example those associated with or expressed by astrocytes, thus modulating the activity of the 19 BUSINESS.30655163.1 393522-004WO (204863) one or more receptors.
  • ARs adenosine receptors
  • the compound activates astrocytes to treat one or more disclosed diseases or conditions.
  • a disclosed compound influences one or more astrocyte functions.
  • the astrocyte function is selected from glutamate uptake, reactive gliosis, swelling, or release of neurotrophic and neurotoxic factors that act to ameliorate metabolic stress and its consequences.
  • the compound is an AR agonist.
  • the purinergic receptor is an A 3 adenosine receptor (A 3 R).
  • the compound is an A3R agonist.
  • the compound is a partial agonist or biased agonist or biased partial agonist, at an A 3 receptor (A 3 R), such as a human A3 receptor (hA3R).
  • the compound acts as an agonist of an A1 adenosine receptor (A1R).
  • the compound is a biased agonist at an A1 and/or A3 receptor. In some embodiments, the compound acts by dual agonism at an A3R and an A1R.
  • the present invention provides a method of treating or ameliorating a brain injury, disease, or condition, such as a brain injury resulting from a TBI or progressive neurodegenerative disorder, in a patient in need thereof, comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the subject has suffered a TBI, concussion, stroke, partial or total spinal cord transection, or malnutrition.
  • the subject has suffered toxic neuropathies, meningoencephalopathies, neurodegeneration caused by a genetic disorder, age-related neurodegeneration, or a vascular disease; or another disease disclosed in US 8,691,775, which is hereby incorporated by reference.
  • the present invention provides a method of treating or ameliorating a brain injury, disease, or condition, such as a brain injury resulting from a TBI or progressive neurodegenerative disorder, in a patient in need thereof, comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same, wherein the compound is an A1R and/or A3R agonist.
  • the compound is a biased agonist, partial agonist, or biased partial agonist at an A 1 receptor. In some embodiments, the compound is a biased agonist, partial agonist, or biased partial agonist at an A3 receptor. In some embodiments, the compound acts by dual agonism at an A 3 R and an A 1 R.
  • the present invention provides a method of promoting or increasing neuroprotection, neurorestoration, or neuroregeneration in a patient suffering from 20 BUSINESS.30655163.1 393522-004WO (204863) a disease or condition, comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the patient is suffering from a neurodegenerative disease or condition.
  • the patient has suffered a TBI or stroke.
  • Traumatic Brain Injuries [00112] Traumatic brain injuries (TBI) are a distressingly common medical condition. There are no approved treatments for TBI, and most TBI patients are discharged from the hospital with no pharmacological treatment. Repetitive TBI such as concussions can trigger age-associated neurodegeneration that results in a range of symptoms and disabilities over decades. TBIs can happen through sports-related injuries, motor vehicle accidents, falls, explosive impacts, physical assaults, etc.
  • Injuries range widely in their complexity and severity, from “mild” concussions with brief alterations in mental status, cognitive difficulties, or loss of consciousness to “severe” with prolonged periods of unconsciousness and/or amnesia after the injury.
  • USCSF and CDC medical intervention
  • the CDC estimates that 1.6 to 3.8 million additional concussion incidents occur in sports and other recreational pursuits annually that do not present to hospital or emergency departments.
  • Approximately 5-10% of athletes will receive a concussion each sport season.
  • TBI is the leading cause of death and disability in children and young adults (CDC) and the most commonly received military-related injury; approximately 20% of U.S. Service Members deployed since 2003 have sustained at least one TBI.
  • Total TBI-related indirect and direct medical costs are estimated at $77 billion annually. At least 5 million Americans require ongoing daily support in performing activities as a result of TBI.
  • a method of treating TBI or promoting recovery from TBI comprising administering to a patient in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the TBI is selected from traumatic injuries to the brain (such as concussion, blast injury, combat-related injury) or spinal cord (such as partial or total spinal cord transection).
  • the TBI results from a mild, moderate, or severe blow to the head, comprises an open or closed head wound, or results from a penetrating or non-penetrating blow to the head.
  • the TBI is selected from 21 BUSINESS.30655163.1 393522-004WO (204863) concussion, blast injury, combat-related injury, a mild, moderate or severe blow to the head, whiplash, sports-related injury, or a head injury sustained from a fall or other accident.
  • Stroke [00114] A stroke occurs when a blood vessel that transports oxygen and nutrients to the brain is disrupted due to an ischemic blockage or from the hemorrhagic rupture of a blood vessel in the brain, causing neurons, glia and endothelial cells in the disrupted region of the brain to die.
  • Strokes can cause unilateral or bilateral paralysis, speech and language disabilities, memory loss, behavioral changes, and even death. Stroke is the fourth leading cause of death in the United States and is a major cause of adult disability. Each year, ⁇ 800,000 people experience a new or recurrent stroke. Each day, over 2,000 Americans will have a stroke, resulting in death in over 400 of these incidents. Stroke accounted for ⁇ 1 of every 19 deaths in the United States in 2010. An estimated 6.8 million Americans ⁇ 20 years of age have had a stroke. As of 2010, the annual direct and indirect cost of stroke was estimated at $36.5 billion.
  • the present invention provides a method of neuroprotective therapy in a stroke patient, comprising administering to a patient in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • such therapy salvages as much of the penumbra as possible, and/or limits further acute tissue damage, and/or promotes neuron recovery.
  • a method of treating stroke or promoting recovery from stroke comprising administering to a patient in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • a method of promoting or increasing neuroprotection, neuroregeneration, or neurorestoration in a patient who has suffered a stroke comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the stroke is selected from ischemic stroke, hemorrhagic stroke, subarachnoid hemorrhage, cerebral vasospasm, and transient ischemic attacks (TIA).
  • the stroke is ischemic, e.g., an acute ischemic stroke (AIS).
  • AIS acute ischemic stroke
  • the stroke is hemorrhagic.
  • the compound is administered within 48 hours of the stroke. In some embodiments, the compound is administered within 24 hours of the stroke. In some embodiments, the compound is administered within 16 hours of the stroke. In some embodiments, the compound is administered within 8, 4, 2, or 1 hours of the stroke.
  • the compound is administered for at least the first 1-72 hours following the stroke. In some embodiments, the compound is administered for at least the first 8-52 hours following the stroke. In some embodiments, the compound is administered for at least the first 8-48 hours following the stroke. In some embodiments, the compound is administered for at least the first 24-48 hours following the stroke. In some embodiments, the compound is administered chronically to treat the stroke as it occurs. In some embodiments, the compound is administered chronically to treat Transient Ischemic Attacks (TIA).
  • TAA Transient Ischemic Attacks
  • the compound is administered chronically to treat ischemic stroke, hemorrhagic stroke, a subarachnoid hemorrhage, cerebral vasospasm, transient ischemic attacks (TIA), or treat a patient who is at an increased risk for a stroke, such as a patient who has had a stroke in the past and is at risk for a further stroke, such as a patient over the age of 40, 45, 50, 55, 60, 65, 70, 75, or 80 years of age.
  • the compound treats an ischemia-reperfusion injury caused by the stroke.
  • a recanalization procedure such as thrombolysis by recombinant tissue plasminogen activator (r- tPA) or mechanical thrombectomy is used in combination with a presently disclosed method of treating stroke or the related condition.
  • r- tPA tissue plasminogen activator
  • Neurodegenerative Diseases are incurable, progressive, and ultimately debilitating syndromes resulting from the progressive degeneration and/or death of neurons in the brain and spinal cord.
  • Neurodegeneration results in movement (ataxias) and/or cognitive function (dementias) disorders, and includes a spectrum of diseases such as Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Huntington’s Disease (HD), Multiple Sclerosis (MS), amyotrophic lateral sclerosis (ALS), and chronic traumatic encephalopathy (CTE). While many 23 BUSINESS.30655163.1 393522-004WO (204863) neurodegenerative diseases are principally genetic in origin, other causes can include viruses, alcoholism, tumors or toxins, and as is now clear, repetitive brain injuries.
  • the present invention provides a method of treating a neurodegenerative disease or promoting recovery from a neurodegenerative disease, comprising administering to a patient in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the present invention provides a method of promoting neuroprotection or neurorestoration in a patient suffering from a neurodegenerative disease, comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • AD Alzheimer’s Disease
  • Activation of astrocytes and promoting neuroprotection and neurorestoration represents a new treatment option for AD.
  • a method of treating AD or promoting neuroprotection or neurorestoration in a patient suffering from AD comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • beneficial effects resulting from a method of treating AD provided herein include, but are not limited to, one or more of: improving cognitive function, 24 BUSINESS.30655163.1 393522-004WO (204863) decreasing cognitive impairment, decreasing plaque burden, enhancing beta amyloid clearance, increasing synaptogenesis, and improving memory.
  • Parkinson’s Disease [00126] As many as one million Americans live with PD, and each year approximately 60,000 Americans are newly diagnosed not including the thousands of cases that go undetected. The total combined direct and indirect cost of PD, including medical treatment, social security payments and lost income, is estimated to be nearly $25 billion per year in the United States. [00127] Activation of neuroprotection and neurorestoration according to the present invention represents a new treatment option for PD. Accordingly, provided herein in one aspect is a method of treating PD or promoting neuroprotection or neurorestoration in a patient suffering from PD, comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • MS Multiple Sclerosis
  • ALS Amyotrophic Lateral Sclerosis
  • Lou Gehrig Lou Gehrig
  • ALS Amyotrophic Lateral Sclerosis
  • Activation of astrocytes can provide stimulation of recovery and repair of the neurons and their connections in an ALS patient.
  • a method of treating ALS or promoting neuroprotection or neurorestoration in a patient suffering from ALS comprising administering to the patient an effective amount of a compound described herein, or a 25 BUSINESS.30655163.1 393522-004WO (204863) pharmaceutically acceptable salt thereof or composition comprising the same.
  • CTE Chronic Traumatic Encephalopathy
  • CTE a form of tauopathy
  • CTE is a progressive neurodegenerative disease found in individuals who have suffered one or more (often multiple or repeated over the course of time) severe blows to the head.
  • CTE is most often diagnosed in professional athletes in American football, soccer, hockey, professional wrestling, stunt performing, bull riding and rodeo performing, motocross, and other contact sports who have experienced brain trauma and/or repeated concussions.
  • CTE chronic traumatic encephalomyopathy
  • ALS progressive muscle weakness and motor and gait abnormalities are believed to be early signs of CTEM.
  • First stage symptoms of CTE include progressive attention deficit, disorientation, dizziness, and headaches.
  • Second stage symptoms comprise memory loss, social instability, erratic behavior, and poor judgment.
  • third and fourth stages patients suffer progressive dementia, slowed movements, tremors, hypomimia, vertigo, speech impediments, hearing loss, and suicidality, and may further include dysarthria, dysphagia, and ocular abnormalities, e.g., ptosis.
  • a method of treating or preventing CTE or promoting neuroprotection or neurorestoration in a patient suffering from CTE comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • a method of stimulating recovery and repair of the neurons and their connections in a CTE patient comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the compound treats one or more symptoms of first stage, second stage, third stage, or fourth stage CTE.
  • the pathology includes neuronal death, tau deposition, TAR DNA-binding Protein 43 (TDP 43) beta-amyloid deposition, white matter changes, and other abnormalities.
  • Tau deposition includes the increasing presence of dense neurofibrillary tangles (NFT), neurites, and glial tangles, which are made up of astrocytes and other glial cells.
  • NFT dense neurofibrillary tangles
  • the method treats, enhances clearance or prevents neuronal death, tau deposition, TAR DNA-binding Protein 43 (TDP 43) beta-amyloid deposition, white matter changes, and other abnormalities associated with CTE.
  • Cardiovascular Diseases are also useful in treating a variety of cardiovascular diseases and conditions.
  • the present invention provides a method of treating a heart (cardiac) or cardiovascular disease, such as cardiac ischemia, myocardial infarction, a cardiomyopathy, chest pain, stress, coronary artery disease, arrhythmia, myocarditis, pericarditis, angina, hypertensive heart disease, endocarditis, rheumatic heart disease, congenital heart disease, or atherosclerosis, comprising administering an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the heart or cardiovascular disease is cardiac ischemia or myocardial infarction.
  • the present invention provides a method of promoting or increasing cardioprotection, cardiorestoration, or cardioregeneration in a patient suffering from a heart (cardiac) or cardiovascular disease or condition, comprising administering to the patient an effective amount of a disclosed compound, or a pharmaceutically acceptable salt thereof or a composition comprising the same.
  • the heart (cardiac) or cardiovascular disease from which the patient is suffering is cardiac ischemia, myocardial infarction, a cardiomyopathy, coronary artery disease, arrhythmia, myocarditis, pericarditis, angina, hypertensive heart disease, endocarditis, rheumatic heart disease, congenital heart disease, or atherosclerosis.
  • cardiac ischemia myocardial infarction
  • coronary artery disease arrhythmia
  • myocarditis myocarditis
  • pericarditis pericarditis
  • angina angina
  • hypertensive heart disease endocarditis
  • rheumatic heart disease congenital heart disease
  • congenital heart disease or atherosclerosis.
  • atherosclerosis congenital heart disease
  • the present invention provides a method of treating neurodegeneration in a patient suffering from a disease or condition, comprising 27 BUSINESS.30655163.1 393522-004WO (204863) administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the present invention provides a method of promoting or increasing neuroprotection, neurorestoration, or neuroregeneration in a patient suffering from a disease or condition, comprising administering to the patient an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the disease or condition is selected from autoimmune diseases, allergic diseases, and/or transplant rejection and graft-versus-host disease (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, WO 2007/20018, hereby incorporated by reference).
  • the disease or condition is selected from intraocular hypertension and/or glaucoma (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, WO 2011/77435, hereby incorporated by reference).
  • the disease or condition is selected from odor sensitivity and/or an olfactory disorder (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, EP1624753, hereby incorporated by reference).
  • the disease or condition is type 2 diabetes (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, US 2010/0256086, hereby incorporated by reference).
  • the disease or condition is selected from respiratory diseases and/or cardiovascular (CV) diseases (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, FASEB J. (2013) 27:1118.4 (abstract of meeting), hereby incorporated by reference).
  • CV cardiovascular
  • the disease or condition is selected from deficits in CNS function, deficits in learning and/or deficits in cognition (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, Neuropsychopharmacology 2015 Jan;40(2):305-14. doi: 10.1038/npp.2014.173. Epub 2014 Jul 15.
  • the disease or condition is selected from a neurodegenerative disease such as Alzheimer's disease, Parkinson’s disease, Huntington’s disease, prion disease, and/or amyotrophic lateral sclerosis (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, US 8,691,775, hereby incorporated by reference).
  • a neurodegenerative disease such as Alzheimer's disease, Parkinson’s disease, Huntington’s disease, prion disease, and/or amyotrophic lateral sclerosis (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, US 8,691,775, hereby incorporated by reference).
  • the disease or condition is selected from otic disorders, Meniere’s disease, endolymphatic hydrops, progressive hearing loss, noise-induced hearing loss, 28 BUSINESS.30655163.1 393522-004WO (204863) dizziness, vertigo, tinnitus, collateral brain damage associated with radiation cancer therapy, and/or migraine treatment (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, US 2009/0306225; UY31779; and US 8,399,018, each of which is hereby incorporated by reference).
  • the disease or condition is selected from pathological sleep perturbations, depression, sleep disorders in the elderly, Parkinson’s disease, Alzheimer’s disease, epilepsy, schizophrenia, and/or symptoms experienced by recovering alcoholics (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, US 2014/0241990, hereby incorporated by reference).
  • the disease or condition is selected from damage to neurons or nerves of the peripheral nervous system during surgery (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, US 8,685,372, hereby incorporated by reference).
  • the disease or condition is a cancer such as prostate cancer (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, Biochem Pharmacol. 2011 August 15; 82(4): 418–425. doi:10.1016/j.bcp.2011.05.013. “Activation of the P2Y1 Receptor Induces Apoptosis and Inhibits Proliferation of Prostate Cancer Cells,” Qiang Wei et al., hereby incorporated by reference).
  • the disease or condition is selected from one or more gastrointestinal conditions such as constipation and/or diarrhea (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, Acta Physiol (Oxf). 2014 Dec;212(4):293-305. doi: 10.1111/apha.12408.
  • gastrointestinal conditions such as constipation and/or diarrhea
  • the disease or condition is selected from cancer of the brain, such as glioblastoma (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, Purinergic Signal. 2015 Sep;11(3):331-46. doi: 10.1007/s11302-015-9454-7. Epub 2015 May 15.
  • the disease or condition is selected from a gastrointestinal disorder such as diarrhea (for the use of certain nucleoside and nucleotide compounds in treating these conditions, see, for example, Acta Physiol (Oxf). 2014 Dec;212(4):293-305. doi: 29 BUSINESS.30655163.1 393522-004WO (204863) 10.1111/apha.12408.
  • the disease or condition is impaired cognition (for the use of certain nucleoside and nucleotide compounds in treating this condition, see, for example, Neuropsychopharmacology. 2015 Jan;40(2):305- 14. doi: 10.1038/npp.2014.173. Epub 2014 Jul 15. “Impaired cognition after stimulation of P2Y1 receptors in the rat medial prefrontal cortex,” Koch H, Bespalov A, Drescher K, Franke H, Krügel U.
  • the present invention provides a method of treating a disease or condition associated with brain injury or a neurodegenerative condition, such as epilepsy, migraine, collateral brain damage associated with radiation cancer therapy, depression, mood or behavioral changes, dementia, erratic behavior, suicidality, tremors, Huntington’s chorea, loss of coordination of movement, deafness, impaired speech, dry eyes, hypomimia, attention deficit, memory loss, cognitive difficulties, vertigo, dysarthria, dysphagia, ocular abnormalities, or disorientation, comprising administering to a patient in need thereof an effective amount of a disclosed compound.
  • a disease or condition associated with brain injury or a neurodegenerative condition such as epilepsy, migraine, collateral brain damage associated with radiation cancer therapy, depression, mood or behavioral changes, dementia, erratic behavior, suicidality, tremors, Huntington’s chorea, loss of coordination of movement, deafness, impaired speech, dry eyes, hypomimia, attention deficit, memory loss, cognitive difficulties, vertigo, dysarth
  • the improvement in cognitive or neurological function is measured as a score increase between about 1% and 20% in the delayed verbal recall task of the revised Wechsler Memory Scale.
  • the improvement in cognitive function may be measured as a score increase between about 10% and 20%, or between about 10% and 15%, or between about 5% and 15%.
  • the present invention provides a method of treating an alcohol-related disorder such as ethanol toxicity, hangover, and neurological and other effects of excessive alcohol consumption.
  • the alcohol-related disorder is selected from Wernicke-Korsakoff Syndrome, alcoholic neuropathy, alcohol withdrawal syndrome, alcoholic cerebellar degeneration, and alcoholic myopathy.
  • GABA gamma- aminobutyric acid
  • glutamate is the primary means of exciting it. Following chronic alcohol exposure, the body decreases (i.e., downregulates) the number or sensitivity of GABA receptors and increases (i.e., upregulates) the number or sensitivity of glutamate receptors in an effort to counterbalance alcohol’s sedative effects.
  • Addictive Disorders Disclosed compounds are also useful in treating addictions, addictive behaviors, behavioral addictions, compulsive disorders and behaviors, and related conditions. [00149] The use of certain compounds in treating such addictions, behaviors, and disorders is described in WO/2019/157317, the contents of which are hereby incorporated by reference. [00150] Cocaine self-administering mice exhibit significantly higher glutamate levels in the VTA (ventral tegmental area) of the brain.
  • the VTA in particular the VTA dopamine neurons, serve several functions in the reward system, motivation, cognition, and drug addiction, and may be the focus of several psychiatric disorders.
  • the elevated glutamate levels appear to be due, at least in part, to loss of glutamate uptake into astrocytes. Without wishing to be bound by theory, it is believed that reduced availability of glutamate has negative effects on astrocyte function and this loss of function affects neuronal activity and drug-seeking behavior. It has now been found that the compounds disclosed herein treat or prevent relapse in addicted individuals, for example by reversing such loss of astrocyte function. Such loss of astrocyte function may be partly due to reduced expression of the glutamate transporter (GLT-1) in astrocytes.
  • GLT-1 glutamate transporter
  • the present invention provides a method of preventing, ameliorating, treating, or promoting recovery from an addiction, addictive behavior, behavioral addiction, brain reward system disorder, compulsive disorder, or related condition, comprising 31 BUSINESS.30655163.1 393522-004WO (204863) administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the addiction is to an addictive substance.
  • the addictive substance is a prescription or recreational drug.
  • the addictive substance is selected from alcohol, nicotine, a stimulant, a cannabinoid agonist, or an opioid agonist.
  • the addictive substance is selected from heroin, ***e, alcohol, an inhalant, an opioid, nicotine, an amphetamine, or a synthetic analog, salt, composition, or combination thereof.
  • the amphetamine is selected from bupropion, cathinone, MDMA, or methamphetamine.
  • the prescription or recreational drug is selected from a cannabinoid agonist or opioid agonist.
  • the addiction is an alcohol or nicotine addiction.
  • the subject is a polydrug abuser.
  • the prescription or recreational drug is selected from ***e, heroin, bupropion, cathinone, MDMA, or methamphetamine morphine, oxycodone, hydromorphone, fentanyl, or a combination thereof.
  • a disclosed compound increases energy metabolism mediated by astrocytes, such as astrocyte mitochondria.
  • the compound reverses loss of glutamate uptake into astrocytes caused by a substance with abuse potential.
  • the compound at least partially reverses the remodeling of the brain reward system caused by the addiction.
  • such effects are mediated by brain or CNS adenosine A 3 receptors, such as astrocyte A 3 R in the VTA; or microglia A 3 R.
  • the present invention provides a method of preventing, ameliorating, treating, or promoting recovery from an addiction, addictive behavior, behavioral addiction brain reward system disorder, compulsive disorder, or related condition by increasing energy metabolism mediated by astrocytes, glia, microglia, neurons, endothelium cells, or other cells of the brain and/or CNS, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the method treats or prevents a relapse of an addiction or addictive behavior in the subject.
  • the subject is addicted to one or more addictive substances such as addictive drugs (drugs having abuse potential).
  • addictive drugs drugs having abuse potential
  • drugs include prescription drugs and recreational drugs such as heroin, ***e, nicotine, or an opioid agonist.
  • the present invention provides a method of treating or preventing withdrawal caused by addiction to one or more addictive substances or drugs, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the compound decreases withdrawal symptoms in an addicted individual in withdrawal.
  • the compound treats withdrawal in an addicted individual in withdrawal.
  • the method further comprises co-administering another drug for treating withdrawal and, optionally, counseling such as psychotherapy.
  • the method further comprises a cognitive behavioral therapy.
  • the method further comprises a digital therapeutic.
  • Digital therapeutics include, for example, reSET or reSET-O (Pear Therapeutics).
  • the present invention provides a method of treating or preventing a relapse of a compulsive disorder or compulsive behavior, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or composition comprising the same.
  • the compulsive disorder is obsessive-compulsive disorder (OCD), Tourette syndrome, trichotillomania, anorexia, bulimia, anxiety disorder, psychosis, or post-traumatic stress disorder.
  • OCD obsessive-compulsive disorder
  • Tourette syndrome trichotillomania
  • anorexia bulimia
  • anxiety disorder psychosis
  • post-traumatic stress disorder e.g., Tourette syndrome, trichotillomania, anorexia, bulimia
  • anxiety disorder e.g., depression, etc.
  • Such disorders are known in the art and include gambling, sex addiction, pornography addiction, eating disorders, spending addiction, rage/anger, workaholism, exercise addiction, risk taking addictions (e.g. kleptomania and pyromania), perfectionism, internet or video game addiction, and compulsive use of electronic devices such as texting and checking social media, to name a few.
  • addictions e.g. kleptomania and pyromania
  • perfectionism e.g. kleptomania and pyromania
  • internet or video game addiction e.g. kleptomania and pyromania
  • compulsive use of electronic devices such as texting and checking social media, to name a few.
  • the term “addiction” includes, unless otherwise specified, physical or psychological dependence on a substance. Addiction may involve withdrawal symptoms or mental or physical distress if the substance is withdrawn.
  • Addiction includes drug liking, drug dependence, habit-formation, neurological and/or synaptic changes, development of brain 33 BUSINESS.30655163.1 393522-004WO (204863) reward system disorders, behavioral changes, or other signs or symptoms of addiction in a subject.
  • the term “addictive drug” or “drug having abuse potential” includes drugs and other substances such as nicotine, whether approved by a regulatory body for treatment of a disease or not, that are known to result in clinical, behavioral, or neurological manifestations of addiction or compulsive behavior.
  • the addictive drug includes nicotine, a cannabinoid agonist, a stimulant, or an opioid agonist.
  • “Addictive substance” refers to addictive drugs as well as other substances of abuse such as alcohol.
  • the present invention provides a method of treating, preventing, promoting recovery from, or ameliorating a pain condition or disorder, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof or pharmaceutical composition thereof.
  • the pain condition or disorder is pain control (pain management, e.g., management of chronic pain).
  • the pain condition or disorder is selected from pain mediated by the CNS, such as neuropathic pain, inflammatory pain, or acute pain.
  • pain mediated by the CNS such as neuropathic pain, inflammatory pain, or acute pain.
  • the pain condition or disorder is migraine.
  • the pain condition or disorder is neuropathic pain, inflammatory pain, or acute pain. See, e.g., Tosh, D.K.; Padia, J.; Salvemini, D.; Jacobson, K.A.
  • the pain condition or disorder is central pain syndrome, peripheral neuropathy, corneal neuropathic pain, post stroke pain, or pain caused by multiple sclerosis.
  • the present invention provides a method of treating pain, comprising administering to a subject in need thereof an effective amount of a disclosed compound, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable composition thereof.
  • the pain is neuropathic pain. In some embodiments, the pain is inflammatory pain. In some embodiments, the pain is acute pain. In some embodiments, the pain is chronic pain. In some embodiments, the pain is nociceptive pain. In some embodiments, the pain is non-inflammatory musculoskeletal pain, fibromyalgia syndrome (FMS), or myofascial pain syndrome (MPS).
  • FMS fibromyalgia syndrome
  • MPS myofascial pain syndrome
  • the pain is selected from musculoskeletal pain, fibromyalgia, myofascial pain, pain during menstruation, pain during osteoarthritis, pain during rheumatoid arthritis, pain during gastrointestinal inflammation, pain during inflammation of the heart muscle, pain during multiple sclerosis, pain during neuritis, pain during AIDS, pain during chemotherapy, tumor pain, headache, chronic pain syndrome (CPS), central pain, trigeminal neuralgia, shingles, stamp pain, phantom limb pain, temporomandibular joint disorder, nerve injury, migraine, post-herpetic neuralgia, neuropathic pain encountered as a consequence of injuries, amputation infections, metabolic disorders or degenerative diseases of the nervous system, neuropathic pain associated with diabetes, pseudesthesia, hypothyroidism, uremia, vitamin deficiencies or alcoholism, acute pain after injuries, postoperative pain, pain during acute gout, and pain from an operation.
  • CPS chronic pain syndrome
  • the musculoskeletal pain is neck and shoulder pain and/or spasms, back pain, sciatica, chest ache, or thigh muscle ache.
  • the pain is, or is associated with, otitis externa (OE), otitis media (OM), mastoiditis, bullous myringitis, eustachian tubal catarrh, labyrinthitis, facial nerve neuritis, temporal bone osteoradionecrosis, mal de debarquement, temporal bone fracture, or temporomandibular joint disease.
  • compositions of this invention may also be present in the compositions of this invention.
  • additional therapeutic agents that are 35 BUSINESS.30655163.1 393522-004WO (204863) normally administered to treat a particular disease, or condition, are known as “appropriate for the disease, or condition, being treated.”
  • the present disclosure provides combination treatments comprising 1) an injectable liquid pharmaceutical composition, comprising: (a) compound A: at a concentration of about 5 mg/mL to about 75 mg/mL; and 2) a second agent to be administered together or separately with the injectable liquid pharmaceutical composition.
  • the second agent is a thromobolytic agent.
  • the thrombolytic agent is alteplase or tenecteplase.
  • the second agent is effective for stroke or traumatic brain injury.
  • additional therapeutic agents that are normally administered to treat that condition, may also be present in the compositions of this invention.
  • a provided compound, or composition thereof is administered in combination with other therapeutic agents, such as tissue plasminogen activators and other thrombolytics, blood thinners, statins, ACE inhibitors, angiotensin II receptor blockers (ARBs), beta blockers, calcium channel blockers or diuretics, to a patient in need thereof.
  • tissue plasminogen activators and other thrombolytics such as a tissue plasminogen activators and other thrombolytics, blood thinners, statins, ACE inhibitors, angiotensin II receptor blockers (ARBs), beta blockers, calcium channel blockers or diuretics
  • the tissue plasminogen activator used in combination with compounds or compositions of the invention include, but are not limited to, alteplase, desmoteplase, reteplase, streptokinase, tenecteplase, urokinase, or combinations of any of the above.
  • the blood thinners used in combination with compounds or compositions of the invention include, but are not limited to, warfarin, heparin, apixabam, clopidogrel, aspirin, rivaroxaban, dabigatran, or combinations of any of the above.
  • statins used in combination with compounds or compositions of the invention include, but are not limited to, atorvastatin, rosuvastatin, fluvastatin, lovastatin, pravastatin, simvastatin and pitavastatin, cerivastatin, mevastatin, or combinations of any of the above.
  • the ACE inhibitors used in combination with compounds or compositions of the invention include, but are not limited to, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, trandolapril benazepril, or combinations of any of the above.
  • the angiotensin II receptor blockers (ARBs) used in combination with compounds or compositions of the invention include, but are not limited to, azilsartan, candesartan, eprosartan, irbesartan, losartan, olmesartan, telmisartan, valsartan, fimasartan, or combinations of any of the above.
  • the beta blockers used in combination with compounds or compositions of the invention include, but are not limited to, atenolol, bisoprolol, betaxolol, carteolol, carvedilol, labetalol, metoprolol, nadolol, nebivolol, oxprenolol, penbutolol, pindolol, propranolol, timolol, or combinations of any of the above.
  • the calcium channel blockers used in combination with compounds or compositions of the invention include, but are not limited to, dihydropyridines: amlodipine, cilnidipine, clevidipine, felodipine, isradipine, lercanidipine, levamlodipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine, diltiazem, verapamil, or combinations of any of the above.
  • dihydropyridines amlodipine, cilnidipine, clevidipine, felodipine, isradipine, lercanidipine, levamlodipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine, diltiazem, verapamil, or combinations of any of the above.
  • the diuretics used in combination with compounds or compositions of the invention include, but are not limited to, loop diuretics, thiazide diuretics, thiazide-like diuretics and potassium-sparing diuretics, or combinations of any of the above.
  • the loop diuretics used in combination with compounds or compositions of the invention include, but are not limited to, bumetanide, ethacrynic acid, furosemide, torsemide, or combinations of any of the above.
  • the thiazide diuretics used in combination with compounds or compositions of the invention include, but are not limited to, epitizide, hydrochlorothiazide and chlorothiazide, bendroflumethiazide, methyclothiazide, polythiazide, or combinations of any of the above.
  • the thiazide-like diuretics used in combination with compounds or compositions of the invention include, but are not limited to, indapamide, chlorthalidone, metolazone, or combinations of any of the above.
  • the potassium-sparing diuretics used in combination with compounds or compositions of the invention include, but are not limited to, amiloride, triamterene, spironolactone, eplerenone, or combinations of any of the above.
  • a provided compound, or composition thereof is administered in combination with a second agent selected from nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroids, muscle relaxants, anti-anxiety drugs, serotonin receptor agonists, antidepressants, anticonvulsants, and corticosteroids.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • corticosteroids corticosteroids
  • muscle relaxants include anti-anxiety drugs, serotonin receptor agonists, antidepressants, anticonvulsants, and corticosteroids.
  • the second agent is selected from opioids, triptans, and COX-2 inhibitors.
  • the second agent is ibuprofen, acetaminophen, aspirin, naproxen, diclofenac, diflunisal, etodolac, fenoprofen, flurbiprofen, indomethacin, ketorolac, mefenamic acid, meloxicam, nabumetone, oxaprozin, piroxicam, sulindac, tolmetin, celecoxib, a triptan, oxycodone, morphine, codeine, hydromorphone, oxymorphone, fentanyl, or sufentanil.
  • the second agent is acetaminophen with codeine (Tylenol #2, #3, #4), buprenorphine, fentanyl transdermal patches, hydrocodone with acetaminophen, hydrocodone with ibuprofen, hydrocodone, hydromorphone, meperidine, methadone, morphine, morphine sustained-release (e.g., MS-Contin®, Avinza®, Kadian®) oxycodone sustained-release (e.g., OxyContin®), Oxycodone with acetaminophen (e.g., Percocet®) oxycodone with aspirin (e.g., Percodan®), oxycodone with ibuprofen (e.g., Combunox®), oxymorphone (e.g., Opana®, Opana ER®), pentazocine, propoxyphene with aspirin, propoxyphene with acetaminophen
  • codeine
  • the second agent is a triptan (serotonin receptor agonist) or other migraine treatment, such as almotriptan (e.g., Axert®), eletriptan (e.g., Relpax®), frovatriptan (e.g., Frova®), naratriptan ((e.g., Amerge®), rizatriptan (e.g., Maxalt®), sumatriptan (e.g., Imitrex®), sumatriptan/naproxen sodium (e.g., Treximet®), zolmitriptan (e.g., Zomig), erenumab (e.g., Aimovig®), or fremanezumab (e.g., Ajovy®), meclizine, dimenhydrinate, ubrogepant (e.g., Ubrelvy®), or botulinum toxin type A (e.g., Boto
  • a provided compound, or composition thereof is administered in combination with a mechanical thrombectomy device, to a patient in need thereof.
  • the mechanical thrombectomy device is a stroke thrombectomy device or a coil embolization device for cerebral aneurysm.
  • such a device includes, but is not limited to, a coil retriever, an aspiration device or a stent retriever.
  • a combination of 2 or more therapeutic agents may be administered together with compounds or compositions of the invention.
  • a combination of 3 or more therapeutic agents may be administered together with compounds or compositions of the invention.
  • Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another, normally within five hours from one another.
  • the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present invention provides a single unit dosage form comprising a compound of the present invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • a provided compound and additional therapeutic agent in those compositions which comprise an additional therapeutic agent as described above
  • 39 BUSINESS.30655163.1 393522-004WO (204863) may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive compound can be administered.
  • compositions which comprise an additional therapeutic agent that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent.
  • a dosage of between about 0.001 - 100 mg/kg body weight/day of the additional therapeutic agent can be administered, or about 0.001 mg/kg to about 500 ⁇ g/kg, or about 0.005 mg/kg to about 250 ⁇ g/kg, or about 0.01 mg/kg to about 100 ⁇ g/kg body weight/day of the additional therapeutic agent can be administered.
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the present invention provides a composition comprising a compound of the present invention and one or more additional therapeutic agents.
  • the therapeutic agent may be administered together with a compound of the present invention or may be administered prior to or following administration of a compound of the present invention. Suitable therapeutic agents are described in further detail below.
  • a compound of the present invention may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours before the therapeutic agent.
  • a compound of the present invention may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours following the therapeutic agent.
  • the present invention provides a medicament comprising at least one compound of the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • BUSINESS.30655163.1 393522-004WO (204863) All features of each of the aspects of the invention apply to all other aspects mutatis mutandis.
  • the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner.
  • EXEMPLIFICATION As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures.
  • Example 1 Synthesis of Compound A NH NH NBoc NBoc N N 20% NaSMe aq. N N Boc O, DMAP, 2-Me-THF N N 10% NH H O, 2-Me-THF N N [00211]
  • the formation of the methylthioether (AST-004-01) used 6 equivalents of sodium thiol methane (NaSMe) under aqueous conditions, at 90-105°C for approximately 72 hours.
  • Step 1 Prior to the reaction, the Step 1 product (AST-004-01) was azeotropically dried to remove residual water in 2-methyl tetrahydrofuran (2-MeTHF).
  • the Boc group in the 9 position was removed by reacting with 10% ammonium hydroxide at 0-5 C to form the di-Boc protected intermediate, AST-004-03.
  • the pH was adjusted to 7-8 hydrochloric acid and the organic portion is separated and then concentrated.
  • N-heptane is added, heated to 60 ⁇ 70°C for 30 to 60 minutes, cooled to 15 ⁇ 25°C for ⁇ 2 to 3 hours and the product was filtered, washed with n-heptane, then dried.
  • the Step 3 product (AST-004-03) can be recrystallized a second time from 2-MeTHF and n-heptane to increase purity of the intermediate if needed.
  • Step 4 utilized the Mitsunobu reaction to couple the methanocarba moiety to the di- Boc protected intermediate (AST-004-03) from Step 3. Equal equivalents of di-Boc protected intermediate (AST-004-03) and methanocarba moiety were charged in tetrahydrofuran (THF) to azeotropically dried to remove residual water. Triphenylphosphine (TPP) and diisopropyl azodicarboxylate (DIAD) were added at 1.5 equivalents each at 15-30°C for approximately 7 hours. A solvent exchange was performed to remove THF and replace it with methyl tert-butyl ether (MTBE).
  • TPP tetrahydrofuran
  • DIAD diisopropyl azodicarboxylate
  • Step 4P used preparative chromatography to purify the Step 4 crude product.
  • Step 4P started by concentrating the crude Step 4 product in dichloromethane. Once concentrated, the solution was diluted with n-heptane for loading on a silica column. Prior to the preparative chromatography, two use-tests were performed to confirm elution time of the desired product (approximately 37 to 55 minutes).
  • the mixture was further cooled between 0°C and10°C for 2 to 3 hours and the solids were filtered and washed with MTBE.
  • the crude TFA salt was stirred for 1 to 2 hours at 15°C-30°C and was cooled between 0°C and10°C for 2 to 3 hours and the solids filtered and washed with MTBE for a second time.
  • Methanol and water for injection were added and the pH is adjusted to pH 8-9 with aqueous sodium hydroxide.
  • the solvent was concentrated and water for injection was added. This solvent mixture was concentrated until methanol is less than 3%.
  • the mixture was heated to 80-90°C for 1 to 2 hours then cooled to 10 to 20°C and crystalized for 1 to 3 hours.
  • Step 5R Recrystallization of AST-004 Drug Substance The crude API was dissolved in 16 volumes of 50/50 methanol and WFI and heated to 50°C to 60°C for 1 to 2 hours. The mixture was cooled to 20°C to 40°C and passed through a bed of diatomacious earth and a capsule filter. The solvent mixture was concentrated under reduced pressure and temperature ⁇ 60°C. The solvent was exchanged with isopropanol until water was less than 3%. The mixture was cooled to 0°C to 10°C for 2 to 3 hours to crystalize the AST-004.
  • Step 5M Recovery of API from Mother Liquor Additional API may be recovered from the mother liquor following a similar process as Step 5 Recrystallization.
  • the mother liquor from the Step 5 Recrystallization was exchanged with isopropanol until water content was ⁇ 3% and concentrated to 5 to 10 solvent volumes under reduced pressure and temperature ⁇ 60°C.
  • the mixture was cooled to 0°C to 5°C for ⁇ 13 hours to crystalize additional AST-004.
  • Example 2 Preparation of an Injectable Composition of Compound A
  • Compound A Injection for intravenous use, contains 125 mg of Compound A active pharmaceutical ingredient in a 5 mL fill volume resulting in a 25 mg mg/mL clear and colorless solution.
  • a 40% PEG 400/water for injection (WFI) (v/v) was added as a vehicle, with a 10 mM phosphate buffer, q.s to apparent pH of 7.8 with either sodium hydroxide or ortho-phosphoric acid.
  • Each vial contained a 0.3 mL overfill to allow for adequate withdrawal of the label volume.
  • Each 6R vial is a clear borosilicate glass with a grey West stopper and a white flip off cap.
  • Compound A Injection is intended to be delivered in a 50 mL admixture of Normal Saline (0.9% NaCl) over a 10-minute infusion.
  • Table 1 contains the composition of Compound A Injection.
  • Example 3 Solubility of Compound A [00220] The solubility of Compound A was determined in common pharmaceutical excipients used as solubility enhancers and determined by HPLC.
  • Air in the Vial Headspace [00222] To address the concern of instability caused by oxygen present in air the headspace, two prototypes used air in the headspace while the other two contained an inert nitrogen overlay headspace. All test between the two configurations were indistinguishable except for BHA quantity which indicted that air being present in the headspace consumed more BHA antioxidant. Since no other differences between air and a nitrogen overlay were observed, air and nitrogen headspaces appear equivocal for Compound A drug product formulations.
  • HPLC Degradants [00226] The ingoing Compound A used had no reportable API-related substances reported above the 0.05% LOQ level. Therefore, all impurities in the drug product were a result of degradation during manufacturing or while on stability. The manufacture of formulation was primarily responsible for all related substances in the drug product with total impurities between 0.44% and 0.46% for the four prototypes. The total impurities appear to decline slightly over time. The largest and primary degradation products (AST-004-020 and AST-004- 021) are the two sulfoxide impurities form during manufacture. Typically, the two sulfoxides 47 BUSINESS.30655163.1 393522-004WO (204863) appeared at approximately the same concentration and remained constant over time or storage conditions.
  • Polyethylene glycols are known to contain peroxides and other oxidizing species (Wu et al 2011, Santos et al 2009, Wasylaschuke et al 2006). Peroxide content in PEG increases during storage with high temperatures and exposure to light (Santos et al 2009). Additionally, the oxidative degradation of PEG ultimately leads to the formation of small reactive molecules such as formaldehyde, acetylaldehyde, formic acid and acetic acid (Hemenway et al 2012). Due of the formation of small organic acids, the pH of the PEG solutions becomes more acidic and is correlated with the degradation of the PEG (Hemenway et al 2012).
  • Table 5 Appearance Stability of Compound A Injection with and Without Antioxidants and With and Without Nitrogen 48 BUSINESS.30655163.1 393522-004WO (204863) Storage Results T est Test A rticle Condition T0 2W 1M 2M 3M s w/ ne s n te n te s w/ ne s n te n te 49 BUSINESS.30655163.1 393522-004WO (204863) Storag Results T est Test e A rticle Condition T0 2W 1M 2M 3M s n te n te n te s n te ne es n te 50 BUSINESS.30655163.1 393522-004WO (204863) est St Results T Test orage A rticle Condition T0 2W 1M 2M 3M n te Additional test results are shown in Tables 6- 9: Table 6: Assay (% of Label Claim) St
  • Table 10 Admixture Stability Protocol for Compound A Injection Time Point (Hours) Storage Conditions T0 2hr 8hr 24hr ted , , .
  • Table 11 Follow-up Admixture Stability Protocol for Compound A Injection Storage Time Point (Hours) Conditions 55 BUSINESS.30655163.1 393522-004WO (204863)
  • Table 12 Admixture Stability and Infusion Set Compatibility Acceptance Criteria Attribute Acceptance Criteria Assay (mg/mL) Report results Compatibility Data
  • Table 13 Low Dose 25mg Admixture Stability and Infusion Set Compatibility of Compound A Injection Results g: H 56 BUSINESS.30655163.1 393522-004WO (204863) Results In Plumb 50 mL 50 mL 50 mL g: H 9) at e L L
  • Table 14 High Dose 375mg Admixture Stability and Infusion Set Compatibility of Compound A Injection Results g: H 57 BUSINESS.30655
  • Preparation for use was performed by transfer of the Compound A Injection to a syringe and then transferred into the saline. After transfer of the Compound A Injection to a 50-mL NaCl 0.9% IV, samples were taken at baseline (T0), 8 and 24 hours at 5°C, and at 8 hours at 25°C. In addition, stability was verified at 2 hours at 25°C for the syringe and infusion set where the admixture was flushed through the infusion set for 5 cycles during the 2 hour period. Testing included visual appearance, color, clarity, visible particles, particulate matter in injections, assay, and purity. For visual appearance, color, and clarity, all results met the predefined acceptance criteria and were identical to the T0 samples.
  • the acceptance criteria were “report results”; however, all results also met the Ph. Eur. 2.9.19 criteria and were not essentially different to the T0 samples.
  • visible particles there were some small white particles in all samples, including the T0 samples and occasionally a fiber.
  • the samples were prepared open to the atmosphere in a non-particle free normal analytical laboratory and as such, some visible particles or fiber were introduced to the samples.
  • pH the ingoing saline had a pH value of 5.0
  • the low dose had a pH between 5.9 and 6.0
  • the high dose had a pH of 6.3.
  • all results met the predefined acceptance criteria and were essentially identical to the T0 samples.
  • Table 16 Batch Formula Table C omponent Function Grade Amount per B atch
  • Compound A Drug Substance was dispensed into a 20L glass vessel and an aqueous buffer solution of disodium phosphate dihydrate and PEG400 was added. The mixture was stirred to mix and dissolve. The pH was checked and 1N sodium hydroxide or ortho- phosphoric acid, 85% was added to adjust to a target pH of 7.8 as necessary.
  • the solution was transferred by peristaltic pump from the first 20L glass vessel to a second 20L glass vessel and pre-filtered using a 0.45 ⁇ m filter.
  • Example 9 Noncompartmental Analysis of Plasma, Urine, and Cerebrospinal Fluid for Compound A for First-In-Human Phase 1 Study in Healthy Subjects
  • the primary study objective was to evaluate the safety and tolerability profile of single ascending IV doses of Compound A in healthy subjects.
  • the secondary study objective 68 BUSINESS.30655163.1 393522-004WO (204863) was to characterize the single dose pharmacokinetic (PK) profile of Compound A in plasma, CSF, and urine.
  • Part I Phase 1 safety, tolerability, and pharmacokinetic (PK) study, conducted in 2 study parts: [00253] Part 1: single ascending dose (SAD); and [00254] Part 2: single dose with cerebrospinal fluid collection (SD CSF). [00255] A total of 40 subjects for Part 1 (SAD) and 12 subjects for Part 2 (SD CSF) were enrolled in the study. The subjects were healthy male and female subjects 18 to 55 years of age at the time of signing the informed consent form. [00256] For Part 1 (SAD), 40 subjects in 5 independent, ascending-dose cohorts were enrolled.
  • SAD single ascending dose
  • SD CSF cerebrospinal fluid collection
  • the doses explored in these 5 cohorts were 5, 25, 50, 75, and 100 mg, which were administered as single 10-minute intravenous (IV) infusions.
  • Each cohort consisted of 8 subjects randomized to Compound A or placebo, whereby 6 subjects received a single dose of Compound A and 2 subjects received placebo.
  • SD CSF For Part 2 (SD CSF), approximately 12 subjects in 1 dose cohort were enrolled who each received a single dose of 100 mg Compound A as a single 10-minute IV infusion (no subjects in SD CSF received a placebo).Sentinel dosing (1 subject to receive Compound A and 1 subject to receive placebo amongst the first 2 subjects) was used in each cohort to ensure adequate safety and tolerability evaluation prior to dosing the remainder of subjects within the cohort.
  • Plasma samples for Part 1 SAD were taken at predose; 10 and 20 minutes; and 0.5, 1, 2, 4, 8, 12, 24, 36, 48, 72, 96, and 120 hours post start of the dose; and optional extended PK timepoints of 144, 168, and 192 hours postdose.
  • Plasma samples for Part 2 SD CSF were taken at predose and 10, 30, 50, and 70 minutes postdose.
  • the complete urine output was collected from each subject during the following intervals: 0 to 6, 6 to 12, 12 to 24, 24 to 36, 36 to 48, 48 to 72, 72 to 96, and 96 to 120 hours postdose.
  • Bioanalytical Analysis The primary analysis variables were the plasma, urine, and CSF concentrations of Compound A, as determined using validated ultra performance liquid chromatography coupled to tandem mass spectrometry (UPLC/MS/MS) bioanalytical assays. The determination of Compound A concentration ranged from 5 to 5000 ng/mL in plasma, 30 to 15,000 ng/mL in urine, and 1 to 500 ng/mL in CSF. The lower limit of quantitation (LLOQ) for the assay used for the quantitation of plasma, urine, and CSF was 5, 30, and 1 ng/mL, respectively.
  • Pharmacokinetic Analysis All exploratory data analyses and presentations of data were performed using RVersion 4.1.3.
  • Noncompartmental analysis modeling was performed using the computer program PKanalix Version 2021R1.
  • AUC0- ⁇ Area under the plasma concentration-time curve from 0 to infinity f ext Percent of AUC 0- ⁇ extrapolated from the last plasma drug concentration to infinity AUMC0- ⁇ Area under the first moment curve from 0 to infinity
  • CL Total plasma clearance calculated as: Dose/AUC 0- ⁇ MRT0- ⁇ Mean residence time from 0 to infinity, calculated as: (AUMC 0- ⁇ /AUC 0- ⁇ )/(D/2), where D is infusion duration Vss Volume of distribution at steady-state, calculated as:
  • Dose Proportionality Assessment Dose proportionality of plasma concentration data was assessed using the power model approach. Dose proportionality was assessed over the dose range of 5 to 100 mg Compound A administered intravenously. The key assumption of this method was that the logarithm of exposure (maximum concentration [Cmax ], area under the plasma concentration-time curve from 0 to the last measurable plasma concentration [AUC0- t], and area under the plasma concentration-time curve from 0 to infinity [AUC0- ⁇ ]) was linearly related to the logarithm of the dose.
  • the calculated point estimate and associated upper and lower confidence interval must be contained wholly within the calculated CI criteria limits.
  • Data Accounting, Inclusion, and Exclusion There were a total of 30 healthy subjects included in the Part 1 SAD analysis, with 6 subjects in each of the dosing cohorts (the 10 placebo subjects were not included in the analyses).
  • One subject randomized to the 5 mg dosing cohort failed to meet the inclusion criteria of negative Hepatitis B surface antigen and was excluded from the PK analyses for plasma and urine, bringing the total number of subjects to 29 for the Part 1 SAD PK analyses. Of the subjects remaining, 1 subject had an outlier observation at 2 hours postdose and another subject had 3 samples collected outside the predetermined sampling windows (deemed as protocol deviations).
  • AUC 0-24 was computed as the cumulative AUC from dose administration to 24 hours after dose.
  • ⁇ AUC 0-t was computed as the cumulative AUC from dose administration to the last observable concentration.
  • ⁇ AUC 0- ⁇ was computed as the cumulative AUC to time to last quantifiable concentration (T last ) of last quantifiable concentration (C last ), plus the extrapolated ⁇ AUC from Tlast to time infinity.
  • AUC tlast- ⁇ The area under the plasma concentration-time curve from the last quantifiable observation to infinity (AUC tlast- ⁇ ) is estimated by dividing the Clast by the first-order elimination rate constant (kel).
  • AUC0- ⁇ was not computed or reported in the following circumstances: ⁇ Data were not amenable to determination of kel; and ⁇ Extrapolated component of AUC0- ⁇ was > 25% of AUC0- ⁇ .
  • Terminal elimination rate constant was computed by linear regression of the terminal linear portion of the log concentration-time data.
  • Terminal elimination t 1/ 2 was initially estimated using the default method in PKanalix. The plots were analyzed by visual inspection to assess the quality of the estimation, and the following constraints were placed on the computation of k el : ⁇ There must have been at least 3 quantifiable concentration-time values included in the regression analysis of the terminal linear phase. ⁇ Adjusted R2 > 0.8 was used. ⁇ Maximum concentration was not used in the determination of kel .
  • Vz The geometric mean values for Vz ranged from 1131.62 to 2056.12 mL/kg.
  • the Vz parameter was observed to be lower for 50 mg dose group when compared to the 25 mg dose group. Additionally, the highest variability was observed in the 5-mg dose group (geometric %CV as 27.88%) followed by 50-mg dose group (geometric %CV as 27.41%) for the Vz parameter.
  • Boxplots comparing dose-normalized parameters for Cmax , AUC0- 24, AUC0- t, and AUC0- ⁇ indicate there was an overlapping of the percentiles between the doses for each dose level, suggesting dose linearity.
  • the geometric mean of CLr ranged from 1.9 to 5.74 mL/min/kg.
  • the percent of unchanged dose recovered and CL r were lowest for 25-mg dose group. This group also had the largest variability compared to the other dose groups for all the computed parameters.
  • the mean (SD) percent of unchanged dose recovered at this collection interval for this particular dosing cohort (25 mg) was 0.003% (0.0055) of the total dose. As the value recovered in urine was ⁇ 1%, and did not seem to impact the overall results, the sample was therefore included in the analysis.

Landscapes

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

Abstract

La présente invention concerne des composés, des compositions et des méthodes d'utilisation de ceux-ci pour le traitement de certains troubles et affections, par exemple des lésions cérébrales telles qu'un accident vasculaire cérébral ou des lésions cérébrales traumatiques.
PCT/US2023/036747 2022-11-03 2023-11-03 Compositions pour administration intraveineuse WO2024097387A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263422292P 2022-11-03 2022-11-03
US63/422,292 2022-11-03
US202363521523P 2023-06-16 2023-06-16
US63/521,523 2023-06-16

Publications (1)

Publication Number Publication Date
WO2024097387A1 true WO2024097387A1 (fr) 2024-05-10

Family

ID=90931307

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/036747 WO2024097387A1 (fr) 2022-11-03 2023-11-03 Compositions pour administration intraveineuse

Country Status (1)

Country Link
WO (1) WO2024097387A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090258850A1 (en) * 2007-08-21 2009-10-15 Frincke James M Stabilized therapeutic compositions and formulations
US20180021363A1 (en) * 2016-04-21 2018-01-25 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
WO2018164513A1 (fr) * 2017-03-09 2018-09-13 씨제이헬스케어 주식회사 Préparation stable contenant du bortézomib et son procédé de préparation
US20210030760A1 (en) * 2018-02-09 2021-02-04 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating addiction and related disorders
US20210077518A1 (en) * 2018-09-26 2021-03-18 Astrocyte Pharmaceuticals, Inc. Polymorphic compounds and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090258850A1 (en) * 2007-08-21 2009-10-15 Frincke James M Stabilized therapeutic compositions and formulations
US20180021363A1 (en) * 2016-04-21 2018-01-25 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
WO2018164513A1 (fr) * 2017-03-09 2018-09-13 씨제이헬스케어 주식회사 Préparation stable contenant du bortézomib et son procédé de préparation
US20210030760A1 (en) * 2018-02-09 2021-02-04 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating addiction and related disorders
US20210077518A1 (en) * 2018-09-26 2021-03-18 Astrocyte Pharmaceuticals, Inc. Polymorphic compounds and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE PUBCHEM 23 June 2018 (2018-06-23), ANONYMOUS: "SCHEMBL19807542", XP093171560, retrieved from PUBCHEM SUBSTANCE Database accession no. 374445831 *

Similar Documents

Publication Publication Date Title
RU2336879C2 (ru) Применение n-десметилклозапина для лечения нейропсихиатрических заболеваний у людей
EP3220891B1 (fr) Formulation sublinguale de riluzole
US20200345734A1 (en) Methods of treating ocular conditions
ES2567041T3 (es) Antagonista del receptor de angiotensina II para el tratamiento de diabetes mellitus en gatos
US20210393615A1 (en) Methods for treating neurological conditions and exposure to nerve agents
JP2023516284A (ja) カンナビジオールの組成物および治療的使用
EP2974728A1 (fr) Agents thérapeutiques pour un dysfonctionnement meibomien
CN112930349A (zh) 多晶形化合物及其用途
Klimek et al. MP-AzeFlu in moderate-to-severe allergic rhinitis: a literature review
Andrabi et al. Ropinirole induces neuroprotection following reperfusion-promoted mitochondrial dysfunction after focal cerebral ischemia in Wistar rats
WO2024097387A1 (fr) Compositions pour administration intraveineuse
EA015483B1 (ru) ПРИМЕНЕНИЕ ИНГИБИТОРА p38 КИНАЗЫ ДЛЯ ЛЕЧЕНИЯ ПСИХИАТРИЧЕСКИХ РАССТРОЙСТВ
ES2731253T3 (es) Un profármaco de 1,1'-(1,6-dioxo-1,6-hexanodiil)bis-d-prolina
EP3880198A1 (fr) Composés activateurs du récepteur d'aryl hydrocarbone (ahr) en tant qu'agents thérapeutiques contre le cancer
US20230414518A1 (en) Rapidly infusing cannabinoid compositions, processes of manufacture, and methods of use
CN110075304B (zh) 一种治疗骨关节炎的药物组合物及其用途
US20040223963A1 (en) Method for treating ischemic stroke with melatonin
KR101994596B1 (ko) 신경정신계 장애의 치료를 위한 조성물 및 방법
WO2024144874A2 (fr) Formes posologiques à dissolution rapide et leurs utilisations
JP2012500248A (ja) 不安障害の治療
JP2022519873A (ja) カンナビノイド受容体アゴニスト及びセリンヒドロラーゼ酵素阻害剤ベースの抗不安治療製品
EP3368532B1 (fr) Dérivé de d-proline comme d'agent d'appauvrissement de sap
CN101432000B (zh) 用于变应性眼病或变应性鼻病的包含三环性***并苯并氮杂*衍生物的预防剂或治疗剂
US20210169848A1 (en) Use of neurokinin-1 antagonists to treat pruritus
Kopach et al. Mitochondrial malfunction mediates impaired cholinergic Ca2+ signalling and submandibular salivary gland dysfunction in diabetes

Legal Events

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

Ref document number: 23886733

Country of ref document: EP

Kind code of ref document: A1