WO2016122832A1 - Compositions, therapeutic and prophylactic methods for treatment of neurodegenerative diseases and brain injuries - Google Patents

Compositions, therapeutic and prophylactic methods for treatment of neurodegenerative diseases and brain injuries Download PDF

Info

Publication number
WO2016122832A1
WO2016122832A1 PCT/US2016/000012 US2016000012W WO2016122832A1 WO 2016122832 A1 WO2016122832 A1 WO 2016122832A1 US 2016000012 W US2016000012 W US 2016000012W WO 2016122832 A1 WO2016122832 A1 WO 2016122832A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
composition
compositions
microglia
pharmaceutically acceptable
Prior art date
Application number
PCT/US2016/000012
Other languages
French (fr)
Inventor
James B. LaVALLE
Original Assignee
Natural Formulations
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 Natural Formulations filed Critical Natural Formulations
Publication of WO2016122832A1 publication Critical patent/WO2016122832A1/en

Links

Classifications

    • 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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • 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
    • 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
    • A61K31/7135Compounds containing heavy metals
    • A61K31/714Cobalamins, e.g. cyanocobalamin, i.e. vitamin B12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • 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

Definitions

  • Microglia are immune cells of the central nervous system (CNS) that normally respond to neuronal damage and remove the damaged cells by CNS.
  • CNS central nervous system
  • microglia Activation of microglia is responsible for an inflammatory process in the central nervous system (CNS) that is believed to play an important role in the pathway leading to neuronal cell death in a number of neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, prion diseases, multiple sclerosis and HIV-dementia. Chronic activation of microglia is believed to cause neuronal damage through the release of potentially cytotoxic molecules such as
  • Traumatic brain injury is a leading cause of injury, death and disability in the United States with over 2.4 million TBIs treated in emergency rooms annually with approximately 50,000 deaths, not including those in the military. (Centers for Disease Control and Prevention, 2009). Those with a high risk for TBI's include athletes and military personnel. Approximately 16.5% of TBIs are associated with sports-related injury.
  • AANS American Association of Neurological Surgeons
  • Complications of TBIs include vascular and neurological damage, increased inflammation and edema of the brain, increased infections, coma, seizures, paralysis, pain, loss of cognitive functions, communication and mood changes, and increased risk of degenerative brain disorders (including Alzheimer's disease and Parkinson's.
  • TBI-related disabilities including long-term cognitive and psychological impairments of moderate to severe disability one-year post accident. Disabilities resulting from a TBI depend upon the severity of the injury, the location of the injury, and the age and general health of the individual. Common disabilities of those with TBIs include cognition problems (thinking, memory, and reasoning), sensory processing deficits (sight, hearing, touch, taste, and smell), communication problems (expression and understanding), and behavior or mental health disturbances (depression, anxiety, mood/personality changes, aggressive behavior, and social inappropriateness). More serious head injuries can result in stupor, coma or vegetative state.
  • TBI total economic cost of TBI in the United States, including direct medical and rehabilitation costs and indirect societal economic costs, is estimated to be approximately $76.5 billion.
  • Intracranial surgery is required for many with TBIs, which comes at a great physical and monetary cost.
  • Most physicians are aware of current treatment protocols for severe TBI, 70-80% of events are classified as mild TBIs and the deficits produced by mild TBI are frequently less often recognized.
  • Current pharmacological treatments for TBIs include hyperosmolar agents, glucocorticoids, anticonvulsants, progesterone, non-steroidal anti-inflammatory drugs (NSAIDs), opiates, sedatives/anti-anxiety agents, and skeletal muscle relaxants.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • opiates sedatives/anti-anxiety agents
  • skeletal muscle relaxants Despite current pharmacological and surgical treatments, an estimated 5.3 million residents in the US are living with TBI-related disabilities, including long- term cognitive and psychological impairments
  • Drug treatment for traumatic brain injury and stroke are generally used for treating symptoms of TBI.
  • Current drug therapies typically have significant side effects and contraindications when compared to the invention, including but not limited to drug-induced nutrient depletions, which can lead to metabolic imbalances in the individual and further complicate recovery and prognosis.
  • the present invention may provide a means to inhibit microglia-mediated inflammation, thereby providing a novel therapeutic approach to the treatment and/or prevention of neurodegenerative diseases.
  • Compostions and methods of the present invention may have reduced side-effect profile and which may be amenable to combination therapy.
  • the brain injury is due to trauma. In other embodiments, the brain injury is due to one of the following causes: falls, chemical/toxins, vehicle-related collisions, stroke, violence, sports injuries, infection, explosive blasts and other combat injuries. There is a great unment need in the area of neurodegenerative diseases and brain injuries (including TBI).
  • the present disclosure is directed to a novel composition comprising a combination of naturally occurring agents.
  • the composition may provide a safe and effective means to prevent and treat such conditions.
  • the composition comprises a combination of two ingredients: ginsenoside Rg3 (Rg3) and nicotinamide riboside (NR), to meet these unmet needs.
  • the source of Rg3 isPanax ginseng, C.A. Meyer, which is a natural source of Rg3.
  • Rg3 has been used in traditional Chinese medicine for over 5000 years. Native Americans also have used another Panax species, Panax quinquefolius or American ginseng, as a traditional medicine.
  • the source of Rg3 is Panax quinquefolius.
  • Current commercial interest of ginseng is based upon the purported benefits of ginseng for general health, including positive effects on the endocrine, cardiovascular, immune, and central nervous systems, prevention of fatigue, oxidative damage, mutagenicity, stimulation of male copulatory behavior in laboratory aniamals, and cancer prevention.
  • Rg3 is an isolated chemical constituent formed by steaming the roots of the plant species Panax, including Panax ginseng, Panax quinquifolius and also Panax notoginseng.
  • the source of Rg3 is any species of Panax.
  • Rg3 helps decrease neuroinflammation in the brain after TBI and also is a neuroprotective agent, helping to decrease the risks of a TBI.
  • Ginsenoside Rg3 is a tetracyclic-triterpene saponin compound found in the plant genus Panax (including Asian or Panax ginseng and American ginseng or Panax quinquifolius), with a molecular weight of 784.13.
  • 20(R)-ginsenoside Rg3 is chemically stable, and insoluble in water, while 20(S)- ginsenoside Rg3 is chemically unstable, and easily dissolvable in water.
  • the form of Rg3 in the composition is the 20(R)-ginsenoside Rg3.
  • the molecular structure is as follows:
  • Ginsenosides are considered the primary pharmacologically active components of ginseng, and appear to be responsible for most of the therapeutic effects on the body. As 20(R)-ginsenoside Rg3 is insoluble in water; the bioavailability of its oral preparations is very low, which greatly restricts the fulfillment of its clinical efficacy.
  • compositions are administered intranasally.
  • One advantage of delivering the composition internasally is. improved clinical efficacy.
  • Rg3 has also been reported to decrease microglial activation, inflammation and neuronal cell apoptosis in neurodegenerative conditions, like Parkinson's and Alzheimer's diseases.
  • Rg3 has been reported in laboratory studies to effectively decrease the oxidative inducible nitric oxide (iNOS), increase macrophage scavenger receptor type A (MSRA), reduce inflammatory cytokine expression and significantly reduce the expression of tissue necrosis factor-alpha (TNF-alpha) in activated microglia (Tian et al, 2009).
  • iNOS oxidative inducible nitric oxide
  • MSRA macrophage scavenger receptor type A
  • TNF-alpha tissue necrosis factor-alpha
  • Rg3 increased the survival rate of neurons exposed to TNF-alpha (Joo et al, 2008).
  • Rg3 has also been reported to attenuate N- methyl-D-aspartate (NMDA) receptor-mediated currents and NMDA-induced neurotoxicity (Kim et al, 2004). Inhibition of L-type Ca(2+) channels by Rg3 may also be another mechanism of ginseng-mediated neuroprotection.
  • NMDA N- methyl-D-aspartate
  • composition further comprisesnicotinamide riboside (NR), with a molecular weight of 255.2.
  • NR nicotinamide riboside
  • NR is a pyridine-nucleoside precursor of the trace nutrient vitamin B3 (niacin) and of NAD+ (nicotinamide adenine dinucleotide), and is produced in mammals (milk) and in yeast (Bieganowski et al. 2004).
  • NR offers similar effects in the body as nicotinic acid, but more pronounced effects on NAD+ production in the central and peripheral nervous systems (Bogan et al, 2008).
  • NR In yeast cells, NR clearly qualifies as a vitamin by having properties of rescuing growth of strains deficient in de novo synthesis, improving Sir2 functions, and utilizing a dedicated transporter (Belenkey et al, 2008; Bieganowski et al, 2004; Bogan et al, 2008; Tempel et al, 2007).
  • NAD+ is an essential compound that enables cells to convert fuel to energy and promotes mitochondrial function, which is an important component in human aging and performance.
  • NR is also neuroprotective, helping decrease the risks of a TBI.
  • the composition further comprisesnicotinamide riboside (NR), with a molecular weight of 255.2, and the structure:
  • Nicotinamide riboside NR along with nicotinic acid and tryptophan, are precursors for
  • NR nicotinamide adenine dinucleotide
  • NAD+ nicotinamide adenine dinucleotide
  • NNKs nicotinamide riboside kinases
  • NAD+ is both a coenzyme for hydride-transfer enzymes and a substrate for NAD+-consuming enzymes, which include ADP-ribose transferases, poly(ADP-ribose) polymerases, cADP-ribose synthases and sirtuins.
  • Sirtuins are the physiological regulators of energy and metabolism (Canto et ' al., 2012; Li et al, 2013; Nogueiras et al, 2012).
  • Sirtulins and NR are reported to have neuroprotective ability (Chi et al, 2013).
  • compositions described herein can be used to treat and prevent conditions caused by microglia-mediated inflammation.
  • Such conditions include without limitation, neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, prion diseases, multiple sclerosis and HIV- dementia.
  • the compositions described herein can be used to treat and prevent brain injuries, including TBI.
  • compositions comprising a combination of ginsenoside Rg3 and nicotinamide riboside, in a pharmaceutically acceptable composition.
  • These compositions are designed to provide an alternative to synthetic pharmaceutical agents that are only used as supportive care and can cause undesirable side effects, such as fatigue, dizziness, confusion and nutrient imbalances. Additionally, in some cases, surgery may also be avoided.
  • the composition is formulated for intranasal administration.
  • the composition can be used for the prevention and treatment of brain injuries, including traumatic brain injury or brain injury due to falls, chemical/toxins, vehicle-related collisions, stroke, violence, sports injuries, infection, explosive blasts and other combat injuries.
  • the composition can be used for preventing and treating neurodenerative diseases, such as Parkinson's disease, Alzheimer's disease, dementia, Huntington's disease, prion diseases, multiple sclerosis, amyotrophic lateral sclerosis (ALS), Lyme disease, and/or HIV-dementia.
  • the composition comprises a combination of 1-8% Rg3 and 92-99% NR w/w.
  • the composition comprises a combination of 2-6% Rg3 and 94-98% NR w/w.
  • the composition comprises a combination of approximately 4% Rg3 and approximately 96% NR w/w.
  • the composition is delivered nasally to deliver a dosage of Rg3 between about 0.05 mg/spray to about 0.5 mg/spray .
  • the composition is delivered nasally to deliver a dosage of Rg3 between about 0.1 mg/spray to about 0.3 mg/spray.
  • the composition is delivered nasally to deliver a dosage of Rg3 of about 0.2 mg/spray.
  • the composition is delivered nasally 1 -6 times per day. In a further embodiment, the composition is delivered nasally 1-4 times per day. In yet another embodiment, the composition is delivered nasally 1-3 times per day. In a further em bodiment, the composition is delivered once a day. Alternatively, the composition is delivered twice a day.
  • the present invention also provides for a method of treating a subject with a brain injury by administerting an effective amount of a composition comprising ginsenoside Rg3 and NR.
  • the method comprises administering to the subject an effective amount of a composition of the invention, or a tautomer, pharmaceutically acceptable salt, solvate, clathrate, or a prodrug of Rg3 and/or NR.
  • Specific routes of administration include intranasal delivery and liposomal tablet delivery.
  • Rg3 and NR may act synergistically to help reduce microglial activation, and inhibit
  • NR neurodegeneration caused by oxidative damage.
  • NR activates mitochondrial enzyme production, which helps to dampen the destruction of neuronal cells and to improve neurogenesis.
  • the compounds of the invention containing reactive functional groups also include protected derivatives thereof.
  • "Protected derivatives” are those compounds in which a reactive site or sites are blocked with one ore more protecting groups.
  • suitable protecting groups for hydroxyl groups include benzyl, methoxymethyl, allyl, trimethylsilyl, tert-butyldimethylsilyl, acetate, and the like.
  • suitable amine protecting groups include
  • suitable thiol protecting groups include benzyl, tert- butyl, acetyl, methoxymethyl and the like.
  • Other suitable protecting groups are well known to those of ordinary skill in the art and include those found in T. W. Greene, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc. 1981.
  • the term "compound(s) of this invention” and similar terms refers to Rg3, NR, and/or a pharmaceutically acceptable salt,' solvate, clathrate, hydrate, polymorph or prodrug thereof, and also include protected derivatives thereof.
  • Rg3 and NR also include pharmaceutically acceptable salts, solvates, clathrates, hydrates, polymorphs and prodrugs thereof.
  • the term "pharmaceutically acceptable carrier” means a suitable vehicle which is pharmaceutically acceptable and can be used to deliver an effective amount of the compounds of this invention.
  • An "effective amount” as used herein means an amount of compounds or compositions of this invention that elicits a desired biological or medicinal response, which includes one or more of the following: (1) preventing a disease, condition or disorder in a subject that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, condition or disorder, (2) delaying onset of a disease, condition or disorder in a subject that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, condition or disorder, (3) treating a disease, condition or disorder in a subject that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder, (4) ameliorating the symptoms of a disease, condition or disorder in a subject that is experiencing or displaying the pathology or symptomatology of the disease,
  • exemplary nasal carriers include saline solutions; alcohols such as ethanol; glycols such as propylene glycol; glycol ethers such as polyethylene glycol and combinations of the foregoing with water and/or one another.
  • alcohols such as ethanol
  • glycols such as propylene glycol
  • glycol ethers such as polyethylene glycol and combinations of the foregoing with water and/or one another.
  • PHARMACEUTICAL SCIENCES 14th edition, 1970.
  • the choice of a suitable nasal carrier in accordance with the present invention will depend on the exact nature of the particular nasal dosage form required.
  • the compounds of this invention may be formulated into a nasal solution (for use as drops or as a spray), a nasal suspension, a nasal ointment, a nasal gel or any other nasal form.
  • Specific nasal dosage forms that are useful in this invention are solutions, suspensions and gels. These dosage forms normally contain a major amount of water (typically purified water) . Minor amounts of other ingredients such as tonicity agents (e.g.
  • compositions of this invention are isotonic and/or buffered to the same pH as blood serum.
  • polymorph means solid crystalline forms of a compound of the present invention or complex thereof. Different polymorphs of the same compound can exhibit different physical, chemical and/or spectroscopic properties. Different physical properties include, but are not limited to stability (e.g., to heat or light), compressibility and density (important in formulation and product manufacturing), and dissolution rates (which can affect bioavailability).
  • Differences in stability can result from changes in chemical reactivity (e.g. , differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph) or mechanical characteristics (e.g., tablets crumble on storage as a kinetically favored polymorph converts to thermodynamically more stable polymorph) or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity).
  • Different physical properties of polymorphs can affect their processing. For example, one polymorph might be more likely to form solvates or might be more difficult to filter or wash free of impurities than another due to, for example, the shape or size distribution of particles of it.
  • the term "hydrate” means a compound of the present invention or a salt thereof, that further includes a stoichiometric or non- stoichiometric amount of water bound by non-covalent intermolecular forces.
  • clathrate means a compound of the present invention or a salt thereof in the form of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule (e.g., a solvent or water) trapped within.
  • spaces e.g., channels
  • guest molecule e.g., a solvent or water
  • prodrug means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound of this invention.
  • Prodrugs may become active upon such reaction under biological conditions, or they may have activity in their unreacted forms.
  • Examples of prodrugs contemplated in this invention include, but are not limited to, analogs or derivatives of Rg3 and NR that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs include derivatives of Rg3 and NR that comprise -NO, -N0 2 , -ONO, or -ON0 2 moieties.
  • Prodrugs can typically be prepared using well-known methods, such as those described by 1 BURGER'S MEDICINAL CHEMISTRY AND DRUG
  • biohydrolyzable amide means an amide, ester, carbamate, carbonate, ureide, or phosphate analogue, respectively, that either: 1) does not destroy the biological activity of the compound and confers upon that compound advantageous properties in vivo, such as improved water solubility, improved circulating half-life in the blood (e.g., because of reduced metabolism of the prodrug), improved uptake, improved duration of action, or improved onset of action; or 2) is itself biologically inactive but is converted in vivo to a biologically active compound.
  • biohydrolyzable amides include, but are not limited to, lower alkyl amides, a-amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides.
  • biohydrolyzable esters include, but are not limited to, lower alkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters.
  • biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, aminoacids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.
  • Neurodegenerative diseaseas that may be treated, ameliorated or prevented with a composition of this invention include without limitation Parkinson's disease, Alzheimer's disease, dementia, Huntington's disease, prion diseases, multiple sclerosis, amyotrophic lateral sclerosis (ALS), Lyme disease, and/or HIV-dementia
  • Brain injuries that may be treated or prevented with a composition of this invention include, without limitation, open and closed head injuries, blunt force trauma, infectious diseases, deceleration injuries, chemical/toxins, hypoxia, tumors, and stroke.
  • the brain injury is due to trauma.
  • the brain injury may be a result of falls, vehicle-related collisions, stroke, violence, sports injuries, explosive blasts, and/or other combat injuries.
  • the compositions of the invention may be delivered in the form of microparticles and nanoparticles. Microparticles preferably possess a weight based mean diameter, number based mean diameter and/or a volume based mean diameter of between about 0.5 ⁇ and about 30 ⁇ , e.g.
  • Nanoparticles preferably possess a weight based mean diameter, number based mean diameter and/or a volume based mean diameter of between about 1 and about 1 ,000 nanometers (nm). In one embodiment, the nanoparticles are between about and about 100 nm, and in another embodiment, less than about 50 nm.
  • weight based mean diameter will be understood by the skilled person to include that the average particle size is characterised and defined from a particle size distribution by weight, i.e. a distribution where the existing fraction (relative amount) in each size class is defined as the weight fraction, as obtained by e.g. sieving (e.g. wet sieving).
  • the term "number based mean diameter” will be understood by the skilled person to include that the average particle size is characterised and defined from a particle size distribution by number, i.e. a distribution where the existing fraction (relative amount) in each size class is defined as the number fraction, as measured by e.g. microscopy.
  • the term "volume based mean diameter” will be understood by the skilled person to include that the average particle size is characterised and defined from a particle size distribution by volume, i.e. a distribution where the existing fraction (relative amount) in each size class is defined as the volume fraction, as measured by e.g. laser diffraction.
  • Micro- and nanoparticles of the active ingredients of this invention may be prepared by any standard technique known to the art, including standard
  • micronisation techniques such as grinding, jet milling, dry milling, wet milling, and precipitation.
  • an air elutriation process may be utilised subsequently to prepare specific size fractions, if desired.
  • compositions of the present invention can be incorporated into liposomes.
  • Liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multilamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic,
  • compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients and the like.
  • the preferred lipids are the phospholipids and phosphatidyl cholines, both natural and synthetic. Methods to form liposomes are well known in the art.
  • the compostions of this invention can be delivered via pulmonary administration.
  • Devices for pulmonary delivery include pressurised metered dose inhalers (pMDI's) and dry powder inhalers (DPI's). In pulmonary administration, the size of the active particles is of great importance in determining the site of the absorption.
  • the particles In order that the particles be carried deep into the lungs, the particles must be very fine, for example having a mass median aerodynamic diameter of less than 10 ⁇ . Particles having aerodynamic diameters greater than 10 ⁇ are likely to impact the walls of the throat and generally do not reach the lung. Particles having aerodynamic diameters in the range of about 5 ⁇ to about 0.5 ⁇ will generally be deposited in the respiratory bronchioles, whereas smaller particles having aerodynamic diameters in the range of about 2 ⁇ to about 0.05 ⁇ are likely to be deposited in the alveoli.
  • compositions of this invention are particulary well suited to be delivered mucosally (for example, (intra)nasally, buccally, sublingually or via pulinary delivery).
  • the blood-brain barrier (BBB) limits the ability of systemically administered therapeutic agents to the central nervous system (CNS). This poses a significant challenge to drug development modalities in treating neurological disorders such as TBI.
  • Mucosal delivery offers a noninvasive and rapid method that targets therapeutics to the CNS, bypassing the BBB and minimizing systemic exposure.
  • the compositions of this invention are delivered intranasally.
  • the nasal cavity is covered by a thin mucosa which is well
  • drugs and other molecules are readily transported across the nasal barrier and directly into blood circulation, without first- pass hepatic and intestinal metabolism. This allows the nasal formulation to reach physiological effects much more readily than when using the systemic route.
  • compositions of this invention are delivered sublingually.
  • the active ingredients may be delivered in the form of microparticles or nanoparticles, or incorporated into liposomes.
  • compositions of this invention are delivered via puliminary administration.
  • the compositions of this invention may be administered on any dosing schedule that produces efficacious results.
  • the compositions of this invention may be administered intranassally once, twice or more times per day, providing a dose of about 0.2 mg Rg3 per spray.
  • Rg3 and NR act synergistically to help decrease neuronal inflammation and improve cellular energy production. Both the Rg3 and NR act at different aspects of neuronal recovery, with the Rg3 limiting immune activation that can lead to neuronal damage and the NR stimulating neurogenesis.
  • Another advantage of the present compositions and methods is improved efficacy given the relative proximity of the site of delivery and the site of injury, in view of the fact that Rg3 is poorly absorbed orally.
  • the present invention is illustrated by the following examples, which are not intended to be limiting in any way.
  • Nicotinamide riboside chloride 1.5 g In a base of :
  • Rg3 powder (API Chemicals, San Antonio, TX) was weighed and mixed with DMSO (supplied by PCCA) and Polysorbate 80. Then sterile water was added and mixed for injection, thus forming the Rg3 base. The rest of the components were then weighed and mixed into the Rg3 base . Benzyl alcohol was added to the mixture and spun overnight (minimum of 12 hours).
  • composition was dispensed in sterile amber nasal spray bottle with metered dose.
  • the final pH 5.5 - 6.0.
  • medium chain triglyceride (MCT) oil may optionally be added for sensitive individuals.
  • Exemplary dose 2 sprays intranasally, 2-4 times daily. Prior to each dose, shake well and refrigerate the composition between doses.
  • Composition 2 is a composition of Composition 2:
  • RG3 Two g of RG3 was added it into a 2000 mL beaker. To the beaker was added 50 mL DMSO and 10 mL polysorbate 80 the solution mixed for approximately an hour. To this solution was added 820 mL water for injection and 30 mL MCT oil (ultra pure) and mixed. Then, 50 g nicotinamide riboside (supplied by Chromadex), 6 g beta cyclodextrin and 2g methylcobalamin B 12 were added and mixed for one hour. During the hour mixing, 9 mL benzyl alcohol was added. The resulting suspension was brought to a final volume by adding 20 mL sterile water to beaker and further mixed overnight under a cover of aluminum foil. The resulting solution was adjusted to pH 8-9 with sodium hydroxide. The final solution was dispensed in 30 mL amber nasal spray glass bottles. EXAMPLE 3
  • a composition was made according to the process of Example 2, using an equivalent molar amont of hydroxycobalamin in place of the methylcobalamin.
  • alpha GPC alpha glycerol phosphorylcholine
  • Medication dosage is typically about 0.2 mg/spray of Rg3 and 5 mg/spray of NR with a normal dose of 0.8 mg total Rg3 and 20 mg NR applied to the nasal cavity.
  • Methylcobalamin or hydroxycobalamin is typically 0.2 mg/spray with a normal dose of 0.8 mg total applied to the nasal cavity.
  • a patient may administer the composition usually once or twice a day, but optionally up to 4 times per day.
  • alpha GPC When present, alpha GPC is present from about 3.75 mg/spray to about 7.5 mg/spray with a normal dose of about 15 to 37.5 mg total applied to the nasal cavity.
  • Patients may experience immediate alertness, improved cognitive ability and be able to process multiple tasks; when previously, patients experienced difficulty completing single tasks. Improvement of "brain fog", ability to focus better and concentrate on a task may also be realized after administration of the compositions of the present invention.
  • the neuroregenerative effects of RG3 may take 3-12 weeks. Patient begins to remember thoughts previously unable to recall.
  • Nicotinamide riboside promotes Sir2 silending and extends lifespan via Nrk and Urhl Pnp l/Meul pathways to NAD+. Cell. 2007; 129(3):473-84.
  • Bogan KL Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu Rev Nutr. 2008; 1 15-30.
  • Nicotinamide riboside a trace nutrient in foods, is a vitamin B3 with effects on energy metabolism and neuroprotection. Curr Opin Clin Nutr Metab Care. 2013;18(6):657-61.
  • the ginsenoside Rg3 has a stimulatory effect on insulin signaling in L6 myotubes. Biochem Biophys Res Commun. 2009 Nov 6;389(l):70-3.

Landscapes

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

Abstract

Disclosed are compositions comprising Rg3 ginsenoside and nicotinamide riboside. These compositions are useful for treating and preventing diseases, conditions and disorders caused by microglia-mediated inflammation, including neurodegenerative diseases and brain injuries.

Description

COMPOSITIONS, THERAPEUTIC AND PROPHYLACTIC METHODS FOR TREATMENT OF NEURODEGENERATIVE DISEASES AND BRAIN INJURIES
RELATED APPLICATIONS
This application claims the benefit of the earlier filed U.S. Provisional Application No. 62/125,754 filed on January 30, 2015. The contents of the provisional application are incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
Microglia are immune cells of the central nervous system (CNS) that normally respond to neuronal damage and remove the damaged cells by
phagocytosis. Activation of microglia is responsible for an inflammatory process in the central nervous system (CNS) that is believed to play an important role in the pathway leading to neuronal cell death in a number of neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, prion diseases, multiple sclerosis and HIV-dementia. Chronic activation of microglia is believed to cause neuronal damage through the release of potentially cytotoxic molecules such as
proinflammatory cytokines, reactive oxygen intermediates, proteinases and complement proteins. Therefore, suppression of microglia-mediated inflammation has been considered as an important strategy in neurodegenerative disease therapy. Traumatic brain injury (TBI) is a leading cause of injury, death and disability in the United States with over 2.4 million TBIs treated in emergency rooms annually with approximately 50,000 deaths, not including those in the military. (Centers for Disease Control and Prevention, 2009). Those with a high risk for TBI's include athletes and military personnel. Approximately 16.5% of TBIs are associated with sports-related injury. According to an American Association of Neurological Surgeons (AANS) study utilizing CPSC data, there were an estimated 446,788 sports-related head injuries treated at US hospital emergency rooms in 2009. The population is generally of a young age at the time of injury, so the possibilities of repeated injuries and long-term disability is of great consequence.
There are also clear military needs to treat traumatic brain injuries. In the wars in Iraq and Afghanistan, the number of blast-related traumatic brain injuries are reported to be as high as 320,000.
Complications of TBIs include vascular and neurological damage, increased inflammation and edema of the brain, increased infections, coma, seizures, paralysis, pain, loss of cognitive functions, communication and mood changes, and increased risk of degenerative brain disorders (including Alzheimer's disease and Parkinson's.
An estimated 5.3 million residents in the US are living with TBI-related disabilities, including long-term cognitive and psychological impairments of moderate to severe disability one-year post accident. Disabilities resulting from a TBI depend upon the severity of the injury, the location of the injury, and the age and general health of the individual. Common disabilities of those with TBIs include cognition problems (thinking, memory, and reasoning), sensory processing deficits (sight, hearing, touch, taste, and smell), communication problems (expression and understanding), and behavior or mental health disturbances (depression, anxiety, mood/personality changes, aggressive behavior, and social inappropriateness). More serious head injuries can result in stupor, coma or vegetative state.
As of 2013, the annual economic cost of TBI in the United States, including direct medical and rehabilitation costs and indirect societal economic costs, is estimated to be approximately $76.5 billion. Intracranial surgery is required for many with TBIs, which comes at a great physical and monetary cost. Although most physicians are aware of current treatment protocols for severe TBI, 70-80% of events are classified as mild TBIs and the deficits produced by mild TBI are frequently less often recognized. Current pharmacological treatments for TBIs include hyperosmolar agents, glucocorticoids, anticonvulsants, progesterone, non-steroidal anti-inflammatory drugs (NSAIDs), opiates, sedatives/anti-anxiety agents, and skeletal muscle relaxants. Despite current pharmacological and surgical treatments, an estimated 5.3 million residents in the US are living with TBI-related disabilities, including long- term cognitive and psychological impairments of moderate to severe disability 1 - year post accident.
Drug treatment for traumatic brain injury and stroke are generally used for treating symptoms of TBI. Current drug therapies typically have significant side effects and contraindications when compared to the invention, including but not limited to drug-induced nutrient depletions, which can lead to metabolic imbalances in the individual and further complicate recovery and prognosis. There is a clear need for more treatment options for TBIs with reduced side effects which may treat brain injury and potentially prevent the development of neurodegenerative diseases.
SUMMARY
The present invention may provide a means to inhibit microglia-mediated inflammation, thereby providing a novel therapeutic approach to the treatment and/or prevention of neurodegenerative diseases. Compostions and methods of the present invention may have reduced side-effect profile and which may be amenable to combination therapy.
In one embodiment, the brain injury is due to trauma. In other embodiments, the brain injury is due to one of the following causes: falls, chemical/toxins, vehicle-related collisions, stroke, violence, sports injuries, infection, explosive blasts and other combat injuries. There is a great unment need in the area of neurodegenerative diseases and brain injuries (including TBI). The present disclosure is directed to a novel composition comprising a combination of naturally occurring agents.The composition may provide a safe and effective means to prevent and treat such conditions. In one embodiment, the composition comprises a combination of two ingredients: ginsenoside Rg3 (Rg3) and nicotinamide riboside (NR), to meet these unmet needs.
In one embodiment, the source of Rg3 isPanax ginseng, C.A. Meyer, which is a natural source of Rg3. Rg3 has been used in traditional Chinese medicine for over 5000 years. Native Americans also have used another Panax species, Panax quinquefolius or American ginseng, as a traditional medicine. In one embodiment, the source of Rg3 is Panax quinquefolius. Current commercial interest of ginseng is based upon the purported benefits of ginseng for general health, including positive effects on the endocrine, cardiovascular, immune, and central nervous systems, prevention of fatigue, oxidative damage, mutagenicity, stimulation of male copulatory behavior in laboratory aniamals, and cancer prevention. Rg3 is an isolated chemical constituent formed by steaming the roots of the plant species Panax, including Panax ginseng, Panax quinquifolius and also Panax notoginseng. In one embodiment, the source of Rg3 is any species of Panax. Rg3 helps decrease neuroinflammation in the brain after TBI and also is a neuroprotective agent, helping to decrease the risks of a TBI.
Ginsenoside Rg3 is a tetracyclic-triterpene saponin compound found in the plant genus Panax (including Asian or Panax ginseng and American ginseng or Panax quinquifolius), with a molecular weight of 784.13. There are two optical isomers for ginsenoside Rg3, ie. 20(R)-ginsenoside Rg3 and 20(S)-ginsenoside Rg3. 20(R)-ginsenoside Rg3 is chemically stable, and insoluble in water, while 20(S)- ginsenoside Rg3 is chemically unstable, and easily dissolvable in water. In one embodiment, the form of Rg3 in the composition is the 20(R)-ginsenoside Rg3. The molecular structure is as follows:
Figure imgf000006_0001
20(R) ginsenoside Rg3
Ginsenosides are considered the primary pharmacologically active components of ginseng, and appear to be responsible for most of the therapeutic effects on the body. As 20(R)-ginsenoside Rg3 is insoluble in water; the bioavailability of its oral preparations is very low, which greatly restricts the fulfillment of its clinical efficacy. In one embodiment of the present invention, compositions are administered intranasally. One advantage of delivering the composition internasally is. improved clinical efficacy.
Laboratory studies have reported that Rg3 helps support neurotransmitter function in the brain (Bao et al, 2005, Mannaa et al, 2006). The ginsenosides seem to decrease excitotoxic and oxidative stress-induced neuronal cell damage, leading to enhanced memory effects.
Rg3 has also been reported to decrease microglial activation, inflammation and neuronal cell apoptosis in neurodegenerative conditions, like Parkinson's and Alzheimer's diseases. Rg3 has been reported in laboratory studies to effectively decrease the oxidative inducible nitric oxide (iNOS), increase macrophage scavenger receptor type A (MSRA), reduce inflammatory cytokine expression and significantly reduce the expression of tissue necrosis factor-alpha (TNF-alpha) in activated microglia (Tian et al, 2009). Rg3 increased the survival rate of neurons exposed to TNF-alpha (Joo et al, 2008). Rg3 has also been reported to attenuate N- methyl-D-aspartate (NMDA) receptor-mediated currents and NMDA-induced neurotoxicity (Kim et al, 2004). Inhibition of L-type Ca(2+) channels by Rg3 may also be another mechanism of ginseng-mediated neuroprotection.
Laboratory studies also support the anxiolytic effects of Rg3 (Kim et al, 2009). The reported mechanism was related to the increase of the storage of hepatic glycogen, and the decrease of the accumulation of metabolites including lactic acid and serum urea nitrogen.
In another embodiment, the composition further comprisesnicotinamide riboside (NR), with a molecular weight of 255.2.
NR is a pyridine-nucleoside precursor of the trace nutrient vitamin B3 (niacin) and of NAD+ (nicotinamide adenine dinucleotide), and is produced in mammals (milk) and in yeast (Bieganowski et al. 2004). NR offers similar effects in the body as nicotinic acid, but more pronounced effects on NAD+ production in the central and peripheral nervous systems (Bogan et al, 2008). In yeast cells, NR clearly qualifies as a vitamin by having properties of rescuing growth of strains deficient in de novo synthesis, improving Sir2 functions, and utilizing a dedicated transporter (Belenkey et al, 2008; Bieganowski et al, 2004; Bogan et al, 2008; Tempel et al, 2007). NAD+ is an essential compound that enables cells to convert fuel to energy and promotes mitochondrial function, which is an important component in human aging and performance. NR is also neuroprotective, helping decrease the risks of a TBI. In another embodiment, the composition further comprisesnicotinamide riboside (NR), with a molecular weight of 255.2, and the structure:
Nicotinamide riboside NR, along with nicotinic acid and tryptophan, are precursors for
nicotinamide adenine dinucleotide (NAD+). NR is metabolized into nicotinamide mononucleotide (NMN) once inside the cell, by a phosphorylation step catalyzed by the nicotinamide riboside kinases (NRKs), which in turn is converted into NAD+. NAD+ is involved in redox activities in the mitochondrial electron transport chain, has been identified as a key regulator of the lifespan-extending effects. NAD+ is both a coenzyme for hydride-transfer enzymes and a substrate for NAD+-consuming enzymes, which include ADP-ribose transferases, poly(ADP-ribose) polymerases, cADP-ribose synthases and sirtuins. Sirtuins are the physiological regulators of energy and metabolism (Canto et'al., 2012; Li et al, 2013; Nogueiras et al, 2012). Sirtulins and NR are reported to have neuroprotective ability (Chi et al, 2013).
In one embodiment of this invention the compositions described herein can be used to treat and prevent conditions caused by microglia-mediated inflammation. Such conditions include without limitation, neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, prion diseases, multiple sclerosis and HIV- dementia. In addition, the compositions described herein can be used to treat and prevent brain injuries, including TBI.
This invention solves the unment needs described above by providing compositions comprising a combination of ginsenoside Rg3 and nicotinamide riboside, in a pharmaceutically acceptable composition. These compositions are designed to provide an alternative to synthetic pharmaceutical agents that are only used as supportive care and can cause undesirable side effects, such as fatigue, dizziness, confusion and nutrient imbalances. Additionally, in some cases, surgery may also be avoided. In one embodiment, the composition is formulated for intranasal administration. In another embodiment, the composition can be used for the prevention and treatment of brain injuries, including traumatic brain injury or brain injury due to falls, chemical/toxins, vehicle-related collisions, stroke, violence, sports injuries, infection, explosive blasts and other combat injuries. In a further embodiment, the composition can be used for preventing and treating neurodenerative diseases, such as Parkinson's disease, Alzheimer's disease, dementia, Huntington's disease, prion diseases, multiple sclerosis, amyotrophic lateral sclerosis (ALS), Lyme disease, and/or HIV-dementia. In one embodiment, the composition comprises a combination of 1-8% Rg3 and 92-99% NR w/w. In a further embodiment, the composition comprises a combination of 2-6% Rg3 and 94-98% NR w/w. In another embodiment, the composition comprises a combination of approximately 4% Rg3 and approximately 96% NR w/w. Although this invention envisions all delivery methods, in a particular embodiment, the composition is delivered nasally to deliver a dosage of Rg3 between about 0.05 mg/spray to about 0.5 mg/spray . In a further embodiment, the composition is delivered nasally to deliver a dosage of Rg3 between about 0.1 mg/spray to about 0.3 mg/spray. In yet another embodiment, the composition is delivered nasally to deliver a dosage of Rg3 of about 0.2 mg/spray.
In one embodiment, the composition is delivered nasally 1 -6 times per day. In a further embodiment, the composition is delivered nasally 1-4 times per day. In yet another embodiment, the composition is delivered nasally 1-3 times per day. In a further em bodiment, the composition is delivered once a day. Alternatively, the composition is delivered twice a day.
The present invention also provides for a method of treating a subject with a brain injury by administerting an effective amount of a composition comprising ginsenoside Rg3 and NR. The method comprises administering to the subject an effective amount of a composition of the invention, or a tautomer, pharmaceutically acceptable salt, solvate, clathrate, or a prodrug of Rg3 and/or NR. Specific routes of administration include intranasal delivery and liposomal tablet delivery.
Without wishing to be bound to theory, the combination of Rg3 and NR may act synergistically to help reduce microglial activation, and inhibit
neurodegeneration caused by oxidative damage. NR activates mitochondrial enzyme production, which helps to dampen the destruction of neuronal cells and to improve neurogenesis.
DETAILED DESCRIPTION OF THE INVENTION
Unless indicated otherwise, the compounds of the invention containing reactive functional groups, such as (without limitation) carboxy, hydroxy, thiol, and amino moieties, also include protected derivatives thereof. "Protected derivatives" are those compounds in which a reactive site or sites are blocked with one ore more protecting groups. Examples of suitable protecting groups for hydroxyl groups include benzyl, methoxymethyl, allyl, trimethylsilyl, tert-butyldimethylsilyl, acetate, and the like. Examples of suitable amine protecting groups include
benzyloxycarbonyl, tert-butoxycarbonyl, tert-butyl, benzyl and fluorenylmethyloxy- carbonyl (Fmoc). Examples of suitable thiol protecting groups include benzyl, tert- butyl, acetyl, methoxymethyl and the like. Other suitable protecting groups are well known to those of ordinary skill in the art and include those found in T. W. Greene, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc. 1981.
As used herein, the term "compound(s) of this invention" and similar terms refers to Rg3, NR, and/or a pharmaceutically acceptable salt,' solvate, clathrate, hydrate, polymorph or prodrug thereof, and also include protected derivatives thereof. The terms Rg3 and NR also include pharmaceutically acceptable salts, solvates, clathrates, hydrates, polymorphs and prodrugs thereof.
As used herein, the term "pharmaceutically acceptable carrier" means a suitable vehicle which is pharmaceutically acceptable and can be used to deliver an effective amount of the compounds of this invention. An "effective amount" as used herein means an amount of compounds or compositions of this invention that elicits a desired biological or medicinal response, which includes one or more of the following: (1) preventing a disease, condition or disorder in a subject that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, condition or disorder, (2) delaying onset of a disease, condition or disorder in a subject that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, condition or disorder, (3) treating a disease, condition or disorder in a subject that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder, (4) ameliorating the symptoms of a disease, condition or disorder in a subject that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder. Compositions intended for intranasal delivery may further comprise a pharmaceutically acceptable nasal carrier. Nasal carriers suitable in accordance with the present invention will be apparent to those skilled in the art of nasal
formulations. Exemplary nasal carriers include saline solutions; alcohols such as ethanol; glycols such as propylene glycol; glycol ethers such as polyethylene glycol and combinations of the foregoing with water and/or one another. For still other examples, reference is made to the text entitled "REMINGTON'S
PHARMACEUTICAL SCIENCES", 14th edition, 1970. The choice of a suitable nasal carrier in accordance with the present invention will depend on the exact nature of the particular nasal dosage form required. For example, the compounds of this invention may be formulated into a nasal solution (for use as drops or as a spray), a nasal suspension, a nasal ointment, a nasal gel or any other nasal form. Specific nasal dosage forms that are useful in this invention are solutions, suspensions and gels. These dosage forms normally contain a major amount of water (typically purified water) . Minor amounts of other ingredients such as tonicity agents (e.g. NaCl), pH adjusters (e.g., a base such as NaOH), emulsifiers or dispersing agents, buffering agents, preservatives, wetting agents and jelling agents (e.g., methylcellulose) may also be present. Specific compositions of this invention are isotonic and/or buffered to the same pH as blood serum. As used herein, the term "polymorph" means solid crystalline forms of a compound of the present invention or complex thereof. Different polymorphs of the same compound can exhibit different physical, chemical and/or spectroscopic properties. Different physical properties include, but are not limited to stability (e.g., to heat or light), compressibility and density (important in formulation and product manufacturing), and dissolution rates (which can affect bioavailability). Differences in stability can result from changes in chemical reactivity (e.g. , differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph) or mechanical characteristics (e.g., tablets crumble on storage as a kinetically favored polymorph converts to thermodynamically more stable polymorph) or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity). Different physical properties of polymorphs can affect their processing. For example, one polymorph might be more likely to form solvates or might be more difficult to filter or wash free of impurities than another due to, for example, the shape or size distribution of particles of it. As used herein, the term "hydrate" means a compound of the present invention or a salt thereof, that further includes a stoichiometric or non- stoichiometric amount of water bound by non-covalent intermolecular forces.
As used herein, the term "clathrate" means a compound of the present invention or a salt thereof in the form of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule (e.g., a solvent or water) trapped within.
As used herein and unless otherwise indicated, the term "prodrug" means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound of this invention. Prodrugs may become active upon such reaction under biological conditions, or they may have activity in their unreacted forms. Examples of prodrugs contemplated in this invention include, but are not limited to, analogs or derivatives of Rg3 and NR that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. Other examples of prodrugs include derivatives of Rg3 and NR that comprise -NO, -N02, -ONO, or -ON02 moieties. Prodrugs can typically be prepared using well-known methods, such as those described by 1 BURGER'S MEDICINAL CHEMISTRY AND DRUG
DISCOVERY (1995) 172-178, 949-982 (Manfred E. Wolff ed., 5th ed). As used herein and unless otherwise indicated, the terms "biohydrolyzable amide", "biohydrolyzable ester", "biohydrolyzable carbamate", "biohydrolyzable carbonate", "biohydrolyzable ureide" and "biohydrolyzable phosphate analogue" mean an amide, ester, carbamate, carbonate, ureide, or phosphate analogue, respectively, that either: 1) does not destroy the biological activity of the compound and confers upon that compound advantageous properties in vivo, such as improved water solubility, improved circulating half-life in the blood (e.g., because of reduced metabolism of the prodrug), improved uptake, improved duration of action, or improved onset of action; or 2) is itself biologically inactive but is converted in vivo to a biologically active compound. Examples of biohydrolyzable amides include, but are not limited to, lower alkyl amides, a-amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides. Examples of biohydrolyzable esters include, but are not limited to, lower alkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters. Examples of biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, aminoacids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.
Neurodegenerative diseaseas that may be treated, ameliorated or prevented with a composition of this invention include without limitation Parkinson's disease, Alzheimer's disease, dementia, Huntington's disease, prion diseases, multiple sclerosis, amyotrophic lateral sclerosis (ALS), Lyme disease, and/or HIV-dementia
Brain injuries that may be treated or prevented with a composition of this invention include, without limitation, open and closed head injuries, blunt force trauma, infectious diseases, deceleration injuries, chemical/toxins, hypoxia, tumors, and stroke. In some embodiments, the brain injury is due to trauma. In other embodiments , the brain injury may be a result of falls, vehicle-related collisions, stroke, violence, sports injuries, explosive blasts, and/or other combat injuries. The compositions of the invention may be delivered in the form of microparticles and nanoparticles. Microparticles preferably possess a weight based mean diameter, number based mean diameter and/or a volume based mean diameter of between about 0.5 μπι and about 30 μπι, e.g. about 15 μιη, such as between about 1 μηι and about 10 μπι. Nanoparticles preferably possess a weight based mean diameter, number based mean diameter and/or a volume based mean diameter of between about 1 and about 1 ,000 nanometers (nm). In one embodiment, the nanoparticles are between about and about 100 nm, and in another embodiment, less than about 50 nm. As used herein, the term "weight based mean diameter" will be understood by the skilled person to include that the average particle size is characterised and defined from a particle size distribution by weight, i.e. a distribution where the existing fraction (relative amount) in each size class is defined as the weight fraction, as obtained by e.g. sieving (e.g. wet sieving). As used herein, the term "number based mean diameter" will be understood by the skilled person to include that the average particle size is characterised and defined from a particle size distribution by number, i.e. a distribution where the existing fraction (relative amount) in each size class is defined as the number fraction, as measured by e.g. microscopy. As used herein, the term "volume based mean diameter" will be understood by the skilled person to include that the average particle size is characterised and defined from a particle size distribution by volume, i.e. a distribution where the existing fraction (relative amount) in each size class is defined as the volume fraction, as measured by e.g. laser diffraction. Micro- and nanoparticles of the active ingredients of this invention may be prepared by any standard technique known to the art, including standard
micronisation techniques, such as grinding, jet milling, dry milling, wet milling, and precipitation. In a further embodiment, an air elutriation process may be utilised subsequently to prepare specific size fractions, if desired.
In one embodiment, the compositions of the present invention can be incorporated into liposomes. Liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multilamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic,
physiologically acceptable and metabolisable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients and the like. The preferred lipids are the phospholipids and phosphatidyl cholines, both natural and synthetic. Methods to form liposomes are well known in the art. In another embodiment, the compostions of this invention can be delivered via pulmonary administration. Devices for pulmonary delivery include pressurised metered dose inhalers (pMDI's) and dry powder inhalers (DPI's). In pulmonary administration, the size of the active particles is of great importance in determining the site of the absorption. In order that the particles be carried deep into the lungs, the particles must be very fine, for example having a mass median aerodynamic diameter of less than 10 μπι. Particles having aerodynamic diameters greater than 10 μηι are likely to impact the walls of the throat and generally do not reach the lung. Particles having aerodynamic diameters in the range of about 5 μιη to about 0.5 μιη will generally be deposited in the respiratory bronchioles, whereas smaller particles having aerodynamic diameters in the range of about 2 μιη to about 0.05 μπι are likely to be deposited in the alveoli.
The compositions of this invention are particulary well suited to be delivered mucosally (for example, (intra)nasally, buccally, sublingually or via pulinary delivery). The blood-brain barrier (BBB) limits the ability of systemically administered therapeutic agents to the central nervous system (CNS). This poses a significant challenge to drug development modalities in treating neurological disorders such as TBI. Mucosal delivery offers a noninvasive and rapid method that targets therapeutics to the CNS, bypassing the BBB and minimizing systemic exposure. In one embodiment, the compositions of this invention are delivered intranasally. The nasal cavity is covered by a thin mucosa which is well
vascularized. Therefore, drugs and other molecules, such as Rg3 and NR, are readily transported across the nasal barrier and directly into blood circulation, without first- pass hepatic and intestinal metabolism. This allows the nasal formulation to reach physiological effects much more readily than when using the systemic route.
(Dhuria SV, Hanson LR, Frey WH 2nd. Intranasal deliver to the central nervous system: mechanisms and experimental considerations. J Pharm Sci.
2010;99(4): 1654-73). In another embodiment, the compositions of this invention are delivered sublingually. In the case of sublingual administration, the active ingredients may be delivered in the form of microparticles or nanoparticles, or incorporated into liposomes. In yet another embodiment, the compositions of this invention are delivered via puliminary administration. The compositions of this invention may be administered on any dosing schedule that produces efficacious results. For example, the compositions of this invention may be administered intranassally once, twice or more times per day, providing a dose of about 0.2 mg Rg3 per spray.
Without wishing to be bound by theory, the inventors believe that the present invention helps support physiological mechanisms in which neural tissue is protected from oxidative stress and chronic inflammation. Rg3 and NR act synergistically to help decrease neuronal inflammation and improve cellular energy production. Both the Rg3 and NR act at different aspects of neuronal recovery, with the Rg3 limiting immune activation that can lead to neuronal damage and the NR stimulating neurogenesis. Another advantage of the present compositions and methods is improved efficacy given the relative proximity of the site of delivery and the site of injury, in view of the fact that Rg3 is poorly absorbed orally. The present invention is illustrated by the following examples, which are not intended to be limiting in any way.
EXEMPLIFICATION The combination of Rg3 ginsenoside with nicotinamide riboside for intranasal administration may be manufactured by any licensed pharmacy by using commercially available materials. EXAMPLE 1 Composition 1 :
Ingredients per 30 mL:
20(R) Rg3 ginsenoside, 90% purity 0.06 g
Nicotinamide riboside chloride 1.5 g In a base of :
Polysorbate 80 NF (Tween 80) 0.3 mL
Dimethyl Sulfoxide 2.0 mL
Hydroxypropyl beta-cyclodextrin powder 1.8 g
Methylcobalamin (sterile) 0.06 g
Water for injection 24.95 mL
Benzyl alcohol NF 0.27 mL
Rg3 powder (API Chemicals, San Antonio, TX) was weighed and mixed with DMSO (supplied by PCCA) and Polysorbate 80. Then sterile water was added and mixed for injection, thus forming the Rg3 base. The rest of the components were then weighed and mixed into the Rg3 base . Benzyl alcohol was added to the mixture and spun overnight (minimum of 12 hours).
The composition was dispensed in sterile amber nasal spray bottle with metered dose. The final pH = 5.5 - 6.0.
To the above mixture, medium chain triglyceride (MCT) oil , 2 mL per 30 mL, may optionally be added for sensitive individuals.
Exemplary dose = 2 sprays intranasally, 2-4 times daily. Prior to each dose, shake well and refrigerate the composition between doses.
EXAMPLE 2
Composition 2:
RG3 2 mg/mL, Methylcobalamin 2 mg/mL,
Nicotinomide Riboside 50 mg/mL
Two g of RG3 was added it into a 2000 mL beaker. To the beaker was added 50 mL DMSO and 10 mL polysorbate 80 the solution mixed for approximately an hour. To this solution was added 820 mL water for injection and 30 mL MCT oil (ultra pure) and mixed. Then, 50 g nicotinamide riboside (supplied by Chromadex), 6 g beta cyclodextrin and 2g methylcobalamin B 12 were added and mixed for one hour. During the hour mixing, 9 mL benzyl alcohol was added. The resulting suspension was brought to a final volume by adding 20 mL sterile water to beaker and further mixed overnight under a cover of aluminum foil. The resulting solution was adjusted to pH 8-9 with sodium hydroxide. The final solution was dispensed in 30 mL amber nasal spray glass bottles. EXAMPLE 3
A composition was made according to the process of Example 2, using an equivalent molar amont of hydroxycobalamin in place of the methylcobalamin.
EXAMPLE 4
Additional composition:
2.25 g of alpha glycerol phosphorylcholine (alpha GPC) was added to to 30 mL bacteriostatic water in beaker and mixed until clear. 15 mL of the alpha GPC 75 mg/mL mixture to 15 mL of Example 2 or Example 3 to prepare:
Rg3/Methylcobalamin (Hydroxycobalamin)/Nicotinamide Riboside/Alpha GPC compositions.
Final concentration of the compositions of this example:
Rg3 1 mg/mL
NR 25 mg/mL
methylcobalamin or hydroxycobalamin 1 mg/mL
alpha GPC 32.5 mg/mL Each nasal spray pump deliver 0.1 mL of mixture and the normal dose is two spray per nostril once or twice a day.
EXAMPLE 5:
Medication dosage is typically about 0.2 mg/spray of Rg3 and 5 mg/spray of NR with a normal dose of 0.8 mg total Rg3 and 20 mg NR applied to the nasal cavity. Methylcobalamin or hydroxycobalamin is typically 0.2 mg/spray with a normal dose of 0.8 mg total applied to the nasal cavity. A patient may administer the composition usually once or twice a day, but optionally up to 4 times per day.
When present, alpha GPC is present from about 3.75 mg/spray to about 7.5 mg/spray with a normal dose of about 15 to 37.5 mg total applied to the nasal cavity. Patients may experience immediate alertness, improved cognitive ability and be able to process multiple tasks; when previously, patients experienced difficulty completing single tasks. Improvement of "brain fog", ability to focus better and concentrate on a task may also be realized after administration of the compositions of the present invention. The neuroregenerative effects of RG3 may take 3-12 weeks. Patient begins to remember thoughts previously unable to recall.
The relevant teachings of all publications cited herein that have not explicitly been incorporated herein by reference, are incorporated herein by reference in their entirety. While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims. All examples are meant to be exemplary" and not intened to limit the scope of this invention in any way.
REFERENCES Bao HY, Zhang J, Yeo SJ, et al. Memory enhancing and neuroprotective effects of selectedginsenosides. Arch Pharm Res. 2005 Mar;28(3):335-42.
Belenky P, Bogan KL, Brenner C. NAD+ metabolism in health and disease. Trends Biochem Sci. 2007;32(l ): 12-9.
Belenky P, Racette FG, Bogan KL, et al. Nicotinamide riboside promotes Sir2 silending and extends lifespan via Nrk and Urhl Pnp l/Meul pathways to NAD+. Cell. 2007; 129(3):473-84.
Bieganowski P, Brenner C. Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss-Handler independent route to NAD+ in fungi and humans. Cells. 1 17;495-502.
Bogan KL, Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu Rev Nutr. 2008; 1 15-30.
Canto C, Houtkooper RH, Pirinen E, et al. The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high fat diet-induced obesity. Cell Metab. 2012; 15(6):838-47.
Chi Y, Sauve AA. Nicotinamide riboside, a trace nutrient in foods, is a vitamin B3 with effects on energy metabolism and neuroprotection. Curr Opin Clin Nutr Metab Care. 2013;18(6):657-61.
Joo SS, Yoo YM, Ahn BW, Nam SY, Kim YB, Hwang KW, Lee do I. Prevention of inflammation-mediated neurotoxicity by Rg3 and its role in microglial activation. Biol Pharm Bull. 2008 Jul;31 (7): 1392-6.
Kim M, Ahn BY, Lee JS, et al. The ginsenoside Rg3 has a stimulatory effect on insulin signaling in L6 myotubes. Biochem Biophys Res Commun. 2009 Nov 6;389(l):70-3.
Kim S, Kim T, Ahn K, Park WK, Nah SY, Rhim H. Ginsenoside Rg3 antagonizes NMDA receptors through a glycine modulatory site in rat cultured hippocampal neurons. Biochem Biophys Res Commun. 2004 Oct 1 ;323(2):416-24.
Li X. SIRT1 and energy metabolism. Acta Biochim Biophys Sin (Shanghai).
2013;45(l ):51 -60. Mannaa F, Abdel-Wahhab MA, Ahmed HH, Park MH. Protective role of Panax ginseng extract standardized with ginsenoside Rg3 against acrylamide-induced neurotoxicity in rats. J Appl Toxicol. 2006 May-Jun;26(3): 1 8-206.
Nogueiras R, Habegger KM, Chaudhary N, et al. Sirtuin 1 and sirtuin 3:
Physiological Regulators of Metabolism. Physiol Rev. 2012;92(3): 1479-1514. Tempel W, Rabeh WM, Bogan KL, et al. Nicotinamide riboside kinase structures reveal new pathways to NAD+. PLoS Biol. 2007;5(10):e263.
Tian J, Zhang S, Li G, Liu Z, Xu B. 20(S)-ginsenoside Rg3, a
neuroprotective agent, inhibits mitochondrial permeability transition pores in rat brain. Phytother Res. 2009 Apr;23(4):486-91.

Claims

CLAIMS What is claimed is:
1. A composition comprising ginsenoside Rg3, or a pharmaceutically
acceptable salt thereof, and nicotinamide riboside, or a pharmaceutically acceptable salt thereof, in a pharmaceutically acceptable carrier.
2. The composition of claim 1, wherein the pharmaceutically acceptable carrier is suitable for intranasal delivery.
3. The composition of claim 2, further comprising methylcobalamin, or a
pharmaceutically acceptable salt thereof.
4. The composition of claim 1 , wherein the pharmaceutically acceptable carrier is suitable for sublingual delivery.
5. A method for treating a subject with a disease, condition or disorder caused by microglia-mediated inflammation comprising the step administering to said subject an effective amount of the composition of claim 1.
6. The method of claim 5, wherein the disease, condition or disorder caused by microglia-mediated inflammation is a neurodegenerative disease.
7. The method of claim 6, wherein the disease, condition or disorder caused by microglia-mediated inflammation is a brain injury.
8. The method of claim 7, in which the brain injury is caused by trauma.
9. A method for preventing a disease, condition or disorder in a subject caused by microglia-mediated inflammation, comprising the step administering to said subject an effective amount of the composition of claim 1.
10. The method of claim 9, wherein the disease, condition or disorder caused by microglia-mediated inflammation is a neurodegenerative disease.
1 1. The method of claim 9, wherein the disease, condition or disorder caused by microglia-mediated inflammation is a brain injury.
12. The method of claim 1 1, in which the brain injury is caused by trauma.
PCT/US2016/000012 2015-01-30 2016-01-28 Compositions, therapeutic and prophylactic methods for treatment of neurodegenerative diseases and brain injuries WO2016122832A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562125754P 2015-01-30 2015-01-30
US62/125,754 2015-01-30

Publications (1)

Publication Number Publication Date
WO2016122832A1 true WO2016122832A1 (en) 2016-08-04

Family

ID=56544152

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/000012 WO2016122832A1 (en) 2015-01-30 2016-01-28 Compositions, therapeutic and prophylactic methods for treatment of neurodegenerative diseases and brain injuries

Country Status (1)

Country Link
WO (1) WO2016122832A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018039207A1 (en) * 2016-08-22 2018-03-01 Elysium Health, Inc. Nicotinamide riboside and pterostilbene compositions and methods for treatment of neurodegenerative disorders
CN113712987A (en) * 2021-07-28 2021-11-30 澳门科技大学 For increasing NAD+Horizontal composition and use thereof
US11286274B2 (en) 2017-06-19 2022-03-29 Mitopower Llc Nicotinamide riboside derivatives and their uses
US11833167B2 (en) 2018-12-17 2023-12-05 Mitopower Llc Nicotinyl riboside compounds and their uses

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080090897A1 (en) * 2006-08-11 2008-04-17 The Johns Hopkins University Compositions and methods for neuroprotectin
US20100272702A1 (en) * 2004-06-04 2010-10-28 Washington University Methods and compositions for treating neuropathies
US20120328526A1 (en) * 2011-06-27 2012-12-27 University Of Maryland, Baltimore Modulation of Nad+ Activity in Neuropathophysiological Conditions and Uses Thereof
WO2014059034A2 (en) * 2012-10-09 2014-04-17 President And Fellows Of Harvard College Nad biosynthesis and precursors for the treatment and prevention of cancer and proliferation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100272702A1 (en) * 2004-06-04 2010-10-28 Washington University Methods and compositions for treating neuropathies
US20080090897A1 (en) * 2006-08-11 2008-04-17 The Johns Hopkins University Compositions and methods for neuroprotectin
US20120328526A1 (en) * 2011-06-27 2012-12-27 University Of Maryland, Baltimore Modulation of Nad+ Activity in Neuropathophysiological Conditions and Uses Thereof
WO2014059034A2 (en) * 2012-10-09 2014-04-17 President And Fellows Of Harvard College Nad biosynthesis and precursors for the treatment and prevention of cancer and proliferation

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018039207A1 (en) * 2016-08-22 2018-03-01 Elysium Health, Inc. Nicotinamide riboside and pterostilbene compositions and methods for treatment of neurodegenerative disorders
US11260069B2 (en) 2016-08-22 2022-03-01 Elysium Health, Inc. Nicotinamide riboside and pterostilbene compositions and methods for treatment of neurodegenerative disorders
AU2017316614B2 (en) * 2016-08-22 2023-06-15 Elysium Health, Inc. Nicotinamide riboside and pterostilbene compositions and methods for treatment of neurodegenerative disorders
US11998561B2 (en) 2016-08-22 2024-06-04 Elysium Health, Inc. Nicotinamide riboside and pterostilbene compositions and methods for treatment of neurodegenerative disorders
US11286274B2 (en) 2017-06-19 2022-03-29 Mitopower Llc Nicotinamide riboside derivatives and their uses
US11833167B2 (en) 2018-12-17 2023-12-05 Mitopower Llc Nicotinyl riboside compounds and their uses
CN113712987A (en) * 2021-07-28 2021-11-30 澳门科技大学 For increasing NAD+Horizontal composition and use thereof
CN113712987B (en) * 2021-07-28 2023-10-31 澳门科技大学 For improving NAD + Horizontal composition and use thereof

Similar Documents

Publication Publication Date Title
Pires et al. Intranasal drug delivery: how, why and what for?
KR101365458B1 (en) Pharmaceutical formulation of apomorphine for buccal administration
US10758479B2 (en) Galenical form for the administration of active ingredients by transmucous means
KR101312311B1 (en) Nasal pharmaceutical formulations and methods of using the same
EP2903619B1 (en) Phosphodiesterase inhibitors for treating taste and smell disorders
CA2726221C (en) Antiallergic marine biopolymers
WO2016122832A1 (en) Compositions, therapeutic and prophylactic methods for treatment of neurodegenerative diseases and brain injuries
EP1824450A2 (en) Compositions for intranasal administration
KR101312331B1 (en) Nasal pharmaceutical formulations and methods of using the same
US20190076378A1 (en) Methods of using dipivefrin
Lyseng-Williamson Fentanyl pectin nasal spray: in breakthrough pain in opioid-tolerant adults with cancer
US8703158B2 (en) Theobromine for the treatment of cough
IL309959A (en) Rapid relief of motor fluctuations in parkinson’s disease
US11903925B2 (en) Compositions for preventing and treating pulmonary injury due to ionizing radiation or cytotoxic drugs
Russo et al. Development and investigation of dry powder inhalers for cystic fibrosis
US8202550B2 (en) Compositions for intranasal administration
US20180104250A1 (en) Formulations for treatment of migraines
AU2013384129B2 (en) Oral suspension for treating eosinophilic esophagitis
GB2420707A (en) Compositions for intranasal administration
US20240123023A1 (en) Pharmaceutical angiotensin-(1-7) compositions in the treatment of (sars)-cov- or (sars)-cov-2-infection related diseases
US20210008010A1 (en) Methods of treating a neurological or psychiatric disorder
WO2022254363A1 (en) Lidocaine or articaine for treating covid-19, autoimmune disease or cytokine storm response
WO2023003593A1 (en) Pulmonary biguanide formulations
WO2014199486A1 (en) Medicine for improving dysphagia
NZ731059B2 (en) An inhalable rapamycin formulation for the treatment of pulmonary hypertension

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: 16743815

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16743815

Country of ref document: EP

Kind code of ref document: A1