EP3043792A2 - Methods of treating fragile x syndrome and related disorders - Google Patents

Methods of treating fragile x syndrome and related disorders

Info

Publication number
EP3043792A2
EP3043792A2 EP14830858.8A EP14830858A EP3043792A2 EP 3043792 A2 EP3043792 A2 EP 3043792A2 EP 14830858 A EP14830858 A EP 14830858A EP 3043792 A2 EP3043792 A2 EP 3043792A2
Authority
EP
European Patent Office
Prior art keywords
metadoxine
mice
treatment
fmrl knockout
administration
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP14830858.8A
Other languages
German (de)
English (en)
French (fr)
Inventor
Yaron DANIELY
Dalia Megiddo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arcturus Therapeutics Ltd
Original Assignee
Alcobra Ltd
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
Priority claimed from US14/038,258 external-priority patent/US20150073023A1/en
Application filed by Alcobra Ltd filed Critical Alcobra Ltd
Publication of EP3043792A2 publication Critical patent/EP3043792A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4415Pyridoxine, i.e. Vitamin B6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4015Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having oxo groups directly attached to the heterocyclic ring, e.g. piracetam, ethosuximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates generally to methods of treating or alleviating a symptom of Fragile X syndrome and related disorders.
  • Fragile X Syndrome is associated with a fragile site expressed as an isochromatid gap in the metaphase chromosome at map position Xq 27.3.
  • Fragile X syndrome is a genetic disorder caused by a mutation in the 5' ⁇ untranslated region of the fragile X mental retardation 1 (FMRl) gene, located on the X chromosome.
  • the mutation that causes FXS is associated with a CGG repeat in the fragile X mental retardation gene FMRl. In most healthy individuals, the total number of CGG repeats ranges from less than 10 to 40, with an average of about 29. In fragile X syndrome, the CGG sequence is repeated from 200 to more than 1 ,000 times.
  • Premutation expansions (55-200 CGG repeats) of the FMRl gene are frequent in the general population, with estimated prevalences of 1 per 259 females and 1 per 812 males.
  • Carriers of the premutation typically have normal IQ, although emotional problems such as anxiety are common.
  • Older male carriers of the premutation (50 years and older) develop progressive intention tremor and ataxia.
  • These movement disorders are frequently accompanied by progressive cognitive and behavioral difficulties, including memory loss, anxiety, and deficits of executive function, reclusive or irritable behavior, and dementia.
  • This disorder has been designated fragile X-associated trernor/ataxia syndrome (FXTAS). Magnetic resonance imaging in subjects with FXTAS reveals increases in T2-weighted signal intensity in the middle cerebellar peduncles and adjacent cerebellar white matter.
  • FXTAS fragile X-associated trernor/ataxia syndrome
  • FXS segregates as an X-linked dominant disorder with reduced penetrance. Either sex when carrying the fragile X mutation may exhibit intellectual disability, which is variable in severity. Children and adults with FXS have varying degrees of intellectual disability or learning disabilities and behavioral and emotional problems, including autistic-like features and tendencies. Young children with FXS often have delays in developmental milestones, such as learning how to sit, walk and talk. Affected children may have frequent tantamis, difficulties in paying attention, frequent seizures (e.g., temporal lobe seizures), are often highly anxious, easily overwhelmed, can have sensory hyperarousal disorder, gastrointestinal disorders, and may have speech problems and unusual behaviors, such as hand flapping and hand biting.
  • seizures e.g., temporal lobe seizures
  • FXS can be diagnosed by an established genetic test performed on a sample (e.g., blood sample, buccal sample) from the subject. The test determines whether a mutation or premutation is present in the FMRl gene of the subject based upon the number of CGG repeats.
  • a sample e.g., blood sample, buccal sample
  • Subjects with FXS can also have autism. About 5% of all children diagnosed with autism have a mutation in the FMRl gene and also have fragile X syndrome (FXS). Autism spectrum disorder (ASD) is seen in approximately 30% of males and 20% of females with FXS, and an additional 30% of FXS individuals display autistic symptoms without having the ASD diagnosis. A lthough intellectual disability is a hallmark feature of FXS, subjects with FXS often display autistic features ranging from shyness, poor eye contact, and social anxiety in mild cases to hand flapping, hand biting and perseverative speech in the severely affected. Subjects with FXS display other symptoms associated with autism such as attention deficit and hyperactivity, seizures, hypersensitivity to sensory stimuli obsessive-compulsive behavior and altered
  • FMRP single protein
  • the invention provides methods of treating or alleviating a symptom of Fragile X Syndrome or a related disorder by administering to a subject in need thereof a composition comprising metadoxine.
  • the symptom is for example, impaired learning or impaired sociability.
  • the subject has Fragile X Syndrome or an Autism Spectrum Disorder.
  • the related disorder is an Autism Spectrum Disorder,
  • a total per day dose-of metadoxine of between 100 - 3000 mg is administered the metadoxine is administered daily, ever ⁇ ' other day or weekly,
  • the metadoxine is administered in one, two, or three dosage forms per day.
  • the metadoxine is administered in a sustained release oral dosage form, wherein the metadoxine is formulated as a combination of slow release and immediate release forms.
  • the slow release form provides for sustained release of the metadoxine for at least 8 hours.
  • the relative proportion of the slow release metadoxine to the immediate release metadoxine is between about-60:40 and 80:20.
  • the relative proportion of the slow release metadoxme to the immediate release metadoxine is about 65 :35.
  • Fig. 1 shows the effect of seven days of once daily intraperitoneal (ip) administration of vehicle (V) or metadoxine (M) (100, 150, or 200 mg/kg) in 2-months oldFmrl knockout ( O) or Wild Type (WT) mice on contextual fear conditioning.
  • Panel A shows the effect of vehicle or 150 mg/kg of metadoxine.
  • Panel B shows the effect of vehicle or 100 mg/kg of metadoxine.
  • Panel C shows the effect of vehicle or 200 mg/kg of metadoxine.
  • Fig. 3 shows the effect of seven days of once daily intraperitoneal administration of vehicle (V) or 150 mg/kg metadoxine (M) on Y-maze spontaneous alternation (Panel A), Y-maze rewarded alternation (Panel B) or Y -maze water maze spatial discrimination (Panel C) in 2- months old Fmrl knockout (KO) or Wild Type (WT) mice.
  • V vehicle
  • M metadoxine
  • the successive alleys of the apparatus presented progressively more anxiogenic environments to explore mice. Movement down the alleys therefore assessed anxiety. In addition, overall activity levels could also be quantitated in the apparatus.
  • Fig. 6 shows the effect of seven days of once daily intraperitoneal administration of vehicle (V) or 150 mg/kg metadoxine (M) on whole brain levels of phosphorylation of ERK (indicative of ERK activity ) (Panel A) and Akt (indicative of Akt activity) (Panel B) in 2-months Fmrl knockout (KO) or Wild Type (WT) mice.
  • V vehicle
  • M metadoxine
  • FIG. 9 shows the effect of once daily ip administration of vehicl e (V) or 150 mg/kg metadoxine (M) for 7 days on whole brain levels of phosphorylation of ERK (Panel A) and Akt (Panel B) in 6 month old Fmrl knockout (KO) or Wild Type (WT) mice.
  • Panel A shows ip and oral treatment with vehicle in Fmrl knockout and Wild Type mice.
  • Panel B shows ip and oral treatment with metadoxine in Wild Type mice.
  • Fig. 12 shows the effect of once daily ip or oral administration (po) of vehicle (V) or metadoxme (M) at 150 or 300 mg/kg for 7 days on lymphocyte biomarkers as assessed using flow cytometry in 2 month old Fmrl knockout (KO) and Wi ld Type (WT) mice.
  • Biomarkers shown are pAkt (Panel A) and pERK (Panel B) in / ⁇ ;;>/ ⁇ / knockout or Wild Type mice.
  • Fig. 13 shows the effect of once daily ip of vehicle (V) or 150 mg/kg metadoxine (M) for 7 days on pERK levels in brain regions of two month old Wild Type ( WT) and Fmrl knockout (KO) mice.
  • Fig. 14 shows the effect of once daily ip of vehicle (V) or 150 mg/kg metadoxine (M) for 7 days on pAkt levels in brain regions of two month old Wi ld Type (WT) and Fmrl knockout (KO) mice.
  • Fig. 15 shows the effect of 5 hour treatment with vehicle (V) or 300 ⁇ metadoxine (M) in vitro on filopodia density (Panel A), length (Panel B), and width (Panel C) in neuronal hippocampal cultures from Fmrl knockout (KO) or Wild Type (WT) mice.
  • the present invention relates to the discovery that metadoxme significantly improves cognitive and social functioning in a valid animal model for the Fragile-X Syndrome.
  • metadoxme significantly improved memory and learning during the contextual fear paradigm in a dose-dependent manner, and the two highest dose levels (150 and 200 mg/kg) fully rescued the Fmrl KO mice learning and memory deficit to a similar extent of the WT mice levels. Furthermore, a significant improvement in memory in the Fmrl KO mice treated with 150 mg kg of metadoxme was found in behavioral tests, such as the T-maze, showing significant improvement in cognitive outcomes. These findings were supplemented by an improved social interaction of KO mice treated with 150 mg/kg of metadoxme. Importantly, improved cognitive executive function, working memory and social interaction following treatment with metadoxme in a valid mouse model of Fragile X correlates with normalization of biochemical markers reflective of neuronal signaling pathways and oxidative stress.
  • Fragile X syndrome is the most widespread single-gene cause of autism and inherited cause of mental retardation among boys.
  • Teen with the FMR1 gene mutation can pass it to their children.
  • Approximately 1 in 4,000 males and 1 in 8,000 females have Fragile X syndrome, according to Centers for Disease Control and Prevention (CDC).
  • Not everyone with the mutation will show signs or symptoms of Fragile X, and disabilities will range from mild to severe as well as physical characteristics such as an elongated face, large or protruding ears, large testes
  • Fragile X results from a change or mutation in the Fragile X Mental Retardation 1 (FMR1) gene, which is found on the X chromosome.
  • FMR1 Fragile X Mental Retardation 1
  • FMRP Fragile X Mental Retardation Protein
  • FXS Fragile X Syndrome
  • Autism spectrum disorders are a group of developmental disabilities that can cause significant social, communication and behavior challenges. People with ASDs handle information in their brain differently than other people.
  • ASDs are "spectrum disorders.” That means ASDs affect each person in different ways, and can range from very mild to severe. People with ASDs share some similar symptoms, such as problems with social interaction. But there are differences in when the symptoms start, how severe they are, and the exact nature of the symptoms. ASDs include Autistic Disorder (also called "classic" autism) , Asperger Syndrome and Pervasive Developmental Disorder,
  • the invention provides methods of treating, preventing or alleviating a sign or symptom of Fragile X Syndrome and/or autism spectrum disorders by administering to a subject a composition comprising metadoxine.
  • Fragile X In general, the signs and symptoms of Fragile X fall into five categories: intelligence and learning; physical, social and emotional, speech and language and sensory disorders commonly associated or sharing features with Fragile X. include for example. Individuals with Fragile X have impaired intellectual functioning, social anxiety, language difficulties and sensitivity to certain sensations. Treatment with metadoxine improves learning and increases sociability in subjects with Fragile X Syndrome.
  • Autism spectrum disorders are commonly associated with individuals with Fragile X syndrome. Signs and symptoms of autism include significant language delays, social and communication challenges, and unusual behaviors and interests. Many people with autistic disorder also have intellectual disability. Individuals with Asperger syndrome usually have some milder symptoms of autistic disorder. For example, they may have social challenges and unusual behaviors and interests. Individuals with Pervasive Developmental Disorder (PDD-NOS) People meet some of the criteria for autistic disorder or Asperger syndrome, but not all, may be diagnosed with PDD-NOS. People with PDD-NOS usually have fewer and milder symptoms than those with autistic disorder. The symptoms might cause only social and communication challenges.
  • PDD-NOS Pervasive Developmental Disorder
  • Metadoxine is an ion-pair between pyrrolidone carboxylate (PCA) and pyridoxine (vitamin B6) with the two compounds linked in a single product by salification.
  • PCA pyrrolidone carboxylate
  • vitamin B6 pyridoxine
  • the pairing with PC A synergistically increases the pharmacological activity of pyridoxine (see, e.g., U.S. Patent 4,313,952).
  • Metadoxme is freely soluble in water and in gastric fluid. Oral absorption of the drug is fast with high bioavailability (60-80%).
  • the half-life of metadoxme in human semm is short (40-60 minutes) without appreciable differences between oral and intravenous administration (Addolorato et al., supra; Lu Yuan et al, Chin. Med. 1 2007 120(2) 160-168).
  • Metadoxme is marketed in several countries as a prescription drug in the form of 500 mg tablets and 300 mg injections. Tablets contain 500 mg of metadoxme, microcrystalline cellulose and magnesium stearate. Ampoules contain 300 mg of metadoxme, sodium
  • metadoxme compositions of the invention e.g., formulated in whole or in part for sustained or controlled release, enable more efficient use of metadoxme in the treatment, prevention and/or alleviation of a sign or symptom of Fragile X syndrome and conditions/disorders related thereto, such as autism spectrum disorders.
  • the metadoxme or acceptable derivative thereof may be formulated for immediate release upon administration to the subject.
  • the metadoxme or acceptable derivative thereof may be formulated for sustained and/or controlled release, and may optionally be formulated to have both immediate release and sustained and/or controlled release
  • metadoxme or a physiologically acceptable derivative thereof is formulated for non-chronic administration.
  • Metadoxme formulations useful in the methods of the present invention described in more detail below.
  • the present invention provides a composition comprising metadoxme or a derivative thereof formulated for sustained and/or controlled release when administered to a subject for improving, treating, preventing and/or alleviating of a sign or symptom of Fragile X syndrome and/or conditions/disorders related thereto, such as autism spectrum disorders.
  • the present invention provides a composition comprising metadoxine or a derivative th ereof wh erein a portion of the metadoxine or derivativ e is formulated for sustained and/or controlled release and a portion of the metadoxine or derivative is formulated for immediate release when administered to a subject for improving, treating , preventing and/or alleviating of a sign or symptom of Fragile X syndrome and/or conditions/disorders related thereto, such as autism spectnim disorders.
  • effective serum levels of the active ingredient are achieved within from about 10 to about 20 or 30 or 40 or 50 or 60, 90 minutes, 2h, 3h, 4b, 5h, 6h, 7h, 8h, 9h, lOh following metadoxme or metadoxine derivative administration. In certain embodiments, effective serum levels of the active ingredient in said subject are achieved within from about 5 to about 20 or 30 or 40 or 50 or 60, 90 minutes, 2h, 3h, 4h, 5h, 6h, 7h, 8h, 9h, lOh following metadoxme or metadoxine derivative administration.
  • effective serum l evels of the active ingredient are achieved within from about 20 to about 20 or 30 or 40 or 50 or 60, 90 minutes, 2h, 3h, 4h, 5h, 6h, 7h, 8h, 9h, I Oh following metadoxine or metadoxine derivative administration. In certain embodiments, effective serum levels of the active ingredient are achieved within about 5, 10, 15, 20, 30, 40, 50 or 60, 90 minutes, 2h, 3h, 4h, 5h, 6h, 7h, 8h, 9h, l Oh.
  • the present inventors have developed innovative approaches for the administration of metadoxine or metadoxine derivative based on enteral (via the digestive tract) and/or parenteral (other routes than digestive tract) routes ( W02G09/004629, the contents of which are incorporated by reference in its entirety). These approaches provide for a rational design of delivery systems with desired properties based on the meticulous selection of the carrier, e.g. appropriate surfactants/co-surfactants composition or micro/nano particles (such as liposomes or nano- liposomes) entrapping the active ingredients, or other additives or excipients, for the delivery system of interest.
  • the carrier e.g. appropriate surfactants/co-surfactants composition or micro/nano particles (such as liposomes or nano- liposomes) entrapping the active ingredients, or other additives or excipients, for the delivery system of interest.
  • the enteral delivery systems may be designed for oral administration (tablets, sachets, lozenges, capsules, gelcaps, drops, or other palatable form) or rectal administration (suppository or (mini) enema form), in addition, the delivery system of interest may be in liquid form, for example a drop solution, syrup. Furthermore, the delivery system of interest may be in form of a beverage or food article.
  • the active ingredient s used by the invention may be comprised in a beverage, particularly soft drinks like juices, nectars, water, sparkling water and other sparkling drinks, shakes, milk shakes and other milk-based drinks, and the like. Liquid preparations may also be in the form of concentrated syrups, for diluting with water or sparkling water.
  • the active ingredient/s may be comprised in food articles, such as snack bars, health bars, biscuits, cookies, sweets, confectionery products, ice creams, ice lollies, and the like.
  • the delivery system may be a food or beverage article comprising a physiologically active pyridoxine derivative, particularly pyridoxol L,2-pyrrolidon-5 carboxylate (metadoxine).
  • a physiologically active pyridoxine derivative particularly pyridoxol L,2-pyrrolidon-5 carboxylate (metadoxine).
  • consumption of the food or beverage article of the invention may lead to achi evement of serum levels of the active ingredient wi thin from about 10 to about 40-60 minutes following consumption thereof Examples may be sweets, chocolate, candies and candy bars, energy bars, ice creams, pastry products and the like.
  • compositions used by the invention are not administered by invasive modes of treatment (i.e., are non-invasive).
  • the metadoxine or metadoxine derivative compositions are not administered by intravenous injection.
  • compositions used by the invention are delivered as a microcrystallme powder or a solution suitable for nebulization; for intravagiiial or intrarectal administration, pessaries, suppositories, creams or foams.
  • a preferred formulation is a formulation for oral administration.
  • Another preferred formulation is for topical administration.
  • Another preferred formulation is for transmucosal administration, sublingual, buccal (absorbed through cheek near gumline) administration, admi istration by inhalation or ocular administration, e.g., in eye drops.
  • the present invention provides deliver ⁇ ' systems for safe delivery of a variety of substances due to their special physico-c hemical features, particularly direct absorption, by non-invasive means, and consequent avoidance of side effects.
  • the delivery systems significantly enhance efficiency and quali ty of metadoxine or metadoxine derivative absorption based on its unique physicoehemicaf features, which enables lower concentrations or amounts of active substance to be delivered to a subject in a biological ly active form.
  • the delivery systems of the invention provide for the direct access of the active substance to the tissues and thus provide immediate or near-immediate effects of metadoxine or metadoxine derivative to the treated subject.
  • the present invention uses a non-invasive pharmaceutical delivery system for the improved administration of a physiologically active pyridoxine, particularly pyridoxol L,2 ⁇ pyrrolidon-5 carboxylate (metadoxine), or a physiological [y acceptable derivative thereof, comprising as the active ingredient said physiologically active pyridoxine in a suitable carrier.
  • a physiologically active pyridoxine particularly pyridoxol L,2 ⁇ pyrrolidon-5 carboxylate (metadoxine)
  • metaldoxine pyridoxol L,2 ⁇ pyrrolidon-5 carboxylate
  • serum levels of the active ingredient are achieved within from about 10 to about 40-60 minutes following administration.
  • the invention employs a non-invasive pharmaceutical delivery system for the improved administration of a physiologically active pyridoxine derivative, particularly pyridoxol L,2-pyrrolidon-5 carboxyiate (metadoxine), for use in improvement of cognitive behavior in a subject in need thereof, comprising as the active ingredient said pyridoxine derivative, in a suitable canier.
  • a physiologically active pyridoxine derivative particularly pyridoxol L,2-pyrrolidon-5 carboxyiate (metadoxine)
  • serum levels of said active ingredient are achieved within from about 10 to about 40-60 minutes following administration.
  • the drug delivery systems employed by the invention may be designed for oral, nasal, ocular, rectal, subcutaneous, transferal, transmueosal, sublingual, buccal or inhalation administration.
  • the drug delivery systems may provide the active substance in a controlled release mode.
  • the drug delivery systems of the invention may further comprise at least one additional pharmaceutically active agent.
  • the deliver ⁇ ' systems used by the invention may generally comprise a buffering agent, an agent that adjusts the osmolality thereof, and optionally, one or more pharmaceutically acceptable carriers, excipients and/or additives as known in the art. Supplementary pharmaceutically acceptable active ingredients can also be incorporated into the compositions.
  • the carrier can be solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • polyol for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents and the like.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic composition is contemplated. It is contemplated that the active agent can be delivered by any pharmaceutically acceptable route and in any pharmaceutically acceptable dosage form.
  • Oral forms include, but are not limited to, tablets, capsules, pills, sachets, lozenges, drops, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. Also included are oral rapid-release, time controlled-release, and delayed-release pharmaceutical dosage forms.
  • the active drug components can be administered in a single dosage form or in separate dosage forms to be administered together or independently.
  • the active drag components can be administered in a mixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as "canier"), materials suitably selected with respect to the intended form of administration.
  • the active drug components can be combined with a non-toxic pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, modified sugars, modified starches, methylcellulose and its derivatives, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, and other reducing and non- reducing sugars, magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl beheiiate, calcium stearate and the like.
  • a non-toxic pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, modified sugars, modified starches, methylcellulose and its derivatives, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, and other reducing and non- reducing sugars, magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl beheiiate, calcium stearate and the like.
  • the active drug components can be combined with non-toxic pharmaceutically acceptable inert earners such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrating agents and coloring and flavoring agents can also be incorporated into the mixture.
  • Stabilizing agents such as antioxidants, propyl gallate, sodium ascorbate, citric acid, calcium metabisulphite, hydroquinone, and 7-hydroxycoumarin can also be added to stabilize the dosage forms.
  • Other suitable compounds can include gelatin, sweeteners, natural and synthetic gums such as acacia, tragacanth, or alginates, carboxymethylcellulose, polyethylene, glycol, waxes and the like.
  • Additional suitable pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, magnesium stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidine, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, poiyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • the pharmaceutically acceptable carrier is magnesium stearate. Additional pharmaceutical excipients commonly accepted and used are found in, for example, Remington's Pharmaceutical
  • solutions in suitabl e oil such as sesame or peanut oil or in aqueous propylene glycol can be employed, as well as sterile aqueous solutions of the corresponding water-soluble salts.
  • aqueous solutions may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal injection purposes.
  • the sterile aqueous media employed are all readily obtainable by standard techniques well-known to those skilled in the art.
  • the delivery sysiem used by this invention may be any suitable delivery sysiem used by this invention.
  • an embodiment of the method of the present invention is to administer the therapeutic compound described herein in a sustained release form.
  • Any controlled or sustained release method known to those of ordinary skill in the art may be used with the compositions and methods of the invention such as those described in Langer, Science 249(4976): 1527-33 ( 1990).
  • Such method comprises administering a sustained-release composition, a suppository, or a coated implantable medical device so that a therapeutically effective dose of the composition of the invention is continuously delivered to a subject of such a method.
  • Sustained release may also be achieved using a patch designed and formulated for the purpose.
  • the composition of the invention may be delivered via a capsule which allows sustained- release of the agent over a period of time.
  • Controlled or sustained-release compositions include formulation in lipophilic depots (e.g., fatty acids, waxes, oils).
  • particulate compositions coated with polymers e.g., poloxamers or poloxamines.
  • Sustained release formulae or devices, or any topical formulations may additionally contain compositions to stabilize the composition or permeate physiological barrier such as skin or mucous membrane.
  • Exemplary additional components may include any physiologically acceptable detergent or solvent such as, for example, dimethylsulfoxide (DMSO).
  • compositions comprising a salt ad duct as defined by the invention formulated as a single dose.
  • Said single dose formulation may be an immediate release formulation, a burst formulation, a prolonged release formulation, a sustained release formulation or any other controlled release formulation known to a person skilled in the art,
  • a composition comprising a salt adduct defined by the invention may be a combined dosage formualtion, wherein different types of formulations are administered to a subject, i.e. any combination of an immediate release formulation, a burst formulation, a prolonged release formulation, a sustained release formulation or any other controlled release formulation known to a person skilled in the art, given either in a single dose or in separate doses given separately, concomitantly or sequentially wherein the gap of time between administration of separate dosages is defined based on the condition and severity of disease or disorder of a subject or the physical condition of said subject.
  • a composition used by the methods of the invention are formulated as combined dosage forms, wherein at least one dosage from of a suit adduct defined by the invention is in an immedi ate release form and at least one dosage form of a salt adduct defined by the invention (being the same or different from the salt adduct formulated in the immediate release formulation) is formulated as a controlled (slow and/or sustained) release formulation.
  • the weight ratio of a salt adduct as defined by the invention comprised in said at least one immediate release formulation and at least one controlled release formulation may be 1 : 1, 1 :2, 2:1 , 3:2, 2:3, 1 :3, 3: 1, 4: 1, 1 :4, 5:2, 2:5, 1 :5, 5: 1 .
  • said at least one immediate release form and at least one controll ed rel ease form of a salt adduct defined above may be administered to a subject separately, concomitantly, sequentially, concurrently, consecutively and so forth.
  • said at least one immediate release form is administered initially.
  • said at least one controlled release formulation is administered initially.
  • the metadoxine or metadoxine derivative in compositions of the invention may be formulated for sustained or controlled release over a period of at least 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 hours. In certain embodiments, the metadoxine or metadoxine derivative in compositions used by the invention may be formulated for sustained or controlled release over a period of about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 hours. In certain
  • the metadoxine or metadoxine derivative in compositions used by the invention may be formulated for sustained or controlled release over a period of between about 0.5 or 1 or 2 or 3 or 4 hours and about 5, 6, 7, 8, 9, 10, 1 1 or 12 hours. In certain embodiments, the metadoxine or metadoxine derivative in compositions used by the invention may be formulated for sustained or controlled release over a period of between about 5 or 6 or 7 or 8 hours and about 9, 10, 11 or 12 hours.
  • the metadoxine or metadoxine derivative in compositions used by the invention may be in immediate, fast of burst release form.
  • the metadoxine or metadoxine derivative in compositions used by the invention may be formulated to release up to 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99, 99.5 or 100% of the total metadoxine or metadoxine derivative in about 0.5, 1, 2, 3, 4, 5, 6, 7 or 8 hours.
  • the metadoxine or metadoxine derivative in compositions used by the invention may be formulated to release not less than 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99, 99.5 or 100% of the total metadoxine or metadoxine derivative in about 0.5, 1 , 2, 3, 4, 5, 6, 7 or 8 hours,
  • the metadoxine or metadoxine derivative in compositions used by the invention may be in a combination of sustained or slow rel ease and immedi ate or fast release forms.
  • the relative proportion of sustained or slow release metadoxine or metadoxine derivative to immediate or fast release metadoxine or metadoxine derivative is, e.g., 1 to 99, 5 to 95, 10 to 90, 15 to 85, 20 to 80, 25 to 75, 30 to 70, 35 to 65, 40 to 60, 45 to 55, 50 to 50, 55 to 45, 60 to 40, 65 to 35, 70 to 30, 75 to 25, 80 to 20, 85 to 15, 90 to 10, 95 to 5, or 99 to 1.
  • a polymeric material is used to sustain or control the release of metadoxine or metadoxine deriv ati ve.
  • the type of polymeric materi al and the amount of which is used has a strong influence on the rate of release of metadoxine or metadoxine deri vati ve from the product of the present invention.
  • polymers include both hydrophobic and hydrophilic polymers.
  • hydrophobic polymers include, but are not limited to, ethyl cellulose and other cellulose derivatives, fats such as glycerol palmito- stereate, beeswax, glycowax, eastorwax, carnaubawax, glycerol monostereate or stearyl alcohol, hydrophobic polyacryl amide derivatives and hydrophobic methacrylic acid derivatives, as well as mixtures of these polymers.
  • Hydrophilic polymers include, but are not limited to, hydrophilic cellulose derivatives such as methyl cellulose, hydroxypropylmethyl cellulose,
  • carboxymethylcellulose and hydroxyethyl methyl-cellulose polyvinyl alcohol polyethylene, polypropylene, polystyrene, poiyacrylamide, ethylene vinyl acetate copolymer, polyacrylate, poly- urethane, polyvinylpyrrolidone, polymemylmethacrylate, polyvinyl acetate, polyhydroxyethyl methacrylate, as well as mixtures of these polymers.
  • any mixture of one or more hydrophobic polymer and one or more hydrophilic polymer could optionally be used.
  • a polymeric material to be used in compositions of or used by the invention is macrocrystalline cellulose such as "AVICEL PH 101" manufactured by FMC BioPolymer's .
  • a polymeric material to be used in compositions of or used by the invention is hydroxypropyl methyl-cellulose such as "METHOLOSE", produced by Shm-Etsu Chemical Co.
  • a polymeric material to be used in compositions of or used by the invention is ethyl cellulose such as "ETHOCELTM", manufactured by The Dow Chemical Company.
  • a polymeric material to be used in compositions of or used by the invention is an acrylic polymer such as "EUDRAGIT RS iM ", produced by Rohm GmbH.
  • a polymeric material to be used in compositions of or used by the invention is a colloidal silicone dioxide such as "AEROSILTM", manufactured by Degussa.
  • a polymeric material to be used in compositions of or used by the invention is a poly( vinyl acetate) such as "KOLLICOAT SR", manufactured by B ASF.
  • a polymeric material to be used in compositions of or used by the invention is an ethyl acetate and vinyl acetate solution such as "DURO-TAK", manufactured by Delasco Dermatologic Lab & Supply, Inc.
  • compositions of or used by the invention comprise or consist essentially of about 50, 100, 150, 200, 250, 300, 400, 500, 600, 700, 800, or 900 mg to about 1000, 1500, 2000, 2500 or 3000 mg metadoxine or metadoxme derivative. In certain embodiments, the compositions of or used by the invention comprise or consist essentially of about 5, 100, 500, oriOOO mg to about 2000, 4000, 10,000, 15,000, or 20,000 mg AVICEL PH 101 m .
  • the compositions of or used by the in vention comprise or consist essentially of about 25, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550 or 600 mg to about 650, 700, 750, 800, 850, 900, 950, 1000, 5000, 10,000, 15,000 or 20,000 mg of a polymeric material.
  • the polymeric material is METHOLOSE, ETHOCEL E10TM or EUDRAGIT R8TM.
  • METHOLOSE comprises or consists essentially of between 1 and 90% of the formulation, preferably between 5 and 70%.
  • ETHOCEL lM comprises or consists essentially of between 1 and 30% of the formulation, preferably between 2 and 20%.
  • EUDRAGiT iM comprises or consists essentially of between 1 and 90% of the formulation, preferably between 5 and 70%.
  • delivery systems of or used by the invention comprise delivery devices.
  • the compositions of or used by the invention are delivered by an osmotic process at a controlled rate such as by an osmotic pump.
  • the system may be constructed by coating an osmotically active agent with a rate controlling semipermeable membrane. This membrane may contain an orifice of critical size through which agent is delivered.
  • the dosage form after coming into contact with aqueous fluids, imbibes water at a rate determined by the fluid permeability of the membrane and osmotic pressure of the core formulation. This osmotic imbibition of water results in formation of a saturated solution of active material within the core, which is dispensed at controlled rate from the deliver ⁇ ' orifice in the membrane.
  • the compositions of or used by the invention are delivered using biodegradable microparticles.
  • the system to prepare microparticles consists of an organic phase comprised of a volatile solvent with dissolved polymer and the material to be encapsulated, emulsified in an aqueous phase
  • the biodegradable polymers that can be used for the microparticle matrix comprises polylactic acid (PLA) or the copolymer of lactic and gly colic acid (PLAGA).
  • PLAGA polylactic acid
  • PLAGA copolymer of lactic and gly colic acid
  • the preparation of or used by the present invention may also contain an absorption enhancer and other optional components.
  • absorption enhancers include, but are not limited to, cyclodextrins, phospholipids, chitosan, DMSO, Tween, Brij, glycocholate, saponin, fusidate and energy based enhancing absorption equipment.
  • Optional components present in the dosage forms include, but are not limited to, diluents, binders, lubricants, surfactants, coloring agents, flavors, buffering agents, preservatives, stabilizing agents and the like.
  • Diluents also termed “fillers” include, for example, dicalcium phosphate dinydrate, calcium sulfate, lactose, cellulose, kaolin, mannitol, sodium chloride, dry starch, hydrolyzed starches, silicon dioxide, colloidal silica, titanium oxide, alumina, talc, micro crystalline cellulose, and powdered sugar.
  • the diluents include, for example, ethanol, sorbitol, glycerol, water and the like.
  • Binders are used to impart cohesive qualities to the formulation.
  • Suitable binder materials include, but are not iimited to, starch (including corn starch and pregelatinzed starch), gelatin, sugars (including sucrose, glucose, dextrose, lactose and sorbitol), polyethylene glycol, waxes, natural and synthetic gums, e.g., acacia, tragacanth, sodium alginate, celluloses, and Veegum, and synthetic polymers such as polyrnethacrylates and polyvinylpyrrolidone.
  • Lubricants are used to facilitate manufacture; examples of suitable lubricants include, for example, magnesium stearate, calcium stearate, stearic acid, glyceryl behenate, and polyethylene glycol.
  • Surfactants may be anionic, eationie, amphoteric or nonionic surface active agents, with anionic surfactants preferred.
  • Suitable anionic surfactants include, but are not limited to, those containing carboxvlate, sulfonate and sulfate ions, associated with cations such as sodium, potassium and ammonium ions.
  • Particularly preferred surfactants include, but are not limited to long alkyl chain sulfonates and alkyl and sulfonates such as sodium dodecylbenzene suifonate; dialkyl sodium suifosuccinates, such as sodium bis-(2-ethylhexyl)-sulfosuccinate; and alkyl sulfates such as sodium lauryl sulfate.
  • Stabilizing agents such as antioxidants, include, but are not limited to, propyl gallate, sodium ascorbate, citric acid, calcium metabisulphite, hydroquinone, and 7-hydroxycoumarin.
  • compositions of or used by the invention may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, preservatives, and the like.
  • compositions of or used by the invention may be used alone or in combination with one or more additional therapeutic agents, for the improvement of cognitive behavior.
  • additional therapeutic agents are: amphetamines, methylphe idate HC1, dexmethylphenidate hydrochloride, atomoxetine, reboxetine, fluoxatine, sertraline, paroxetine, fluoroxamine, citalopram, venlafaxine, bupropion, nefazodone and mirtazapine.
  • compositions of this invention should be formulated so that a dosage of between 0.1-1 g/ ' kg body weight/day, preferably 0.1-300 mg/kg body weight, can be administered.
  • the dose of the compound depends on the condition and the illness of the patient, and the desired daily dose. In human therapy, the oral daily dose is 10-3000 rng or preferably 100-3000 rag.
  • the daily dose is 10, 25, 50 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900, 1000, 1 100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, or 3000 mg.
  • These doses are administered in unit dosage forms, which may be administered in a single daily dose or divided into 2-3 smaller doses for each day in certam cases.
  • compositions of the present invention may act
  • the amount of compound(s) and additional therapeutic agent(s) in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent.
  • a dosage of between 0.1-1 g kg bodyweight/ ' day of the additional therapeutic agent can be administered.
  • prophylactic or therapeutic treatment refers to administration to a subject of one or more of the compositions of the invention. If it is administered prior to clinical manifestation of the unwanted condition (e.g., clinical or other unwanted state of the host animal) then the treatment is prophylactic, i.e., it contributes to prevention of, i.e., protection of the subject against developing an unwanted condition, whereas if admmistered after manifestation of an unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate or prevent progression of the unwanted condition or side effects there from).
  • the unwanted condition e.g., clinical or other unwanted state of the host animal
  • the term "therapeutic effect” refers to a local or systemic effect in animals, particularly mammals, and more particularly humans, caused by a pharmacolog cal!' active substance or substances.
  • the term thus means any substance intended for use in diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and conditions in an animal or human.
  • therapeutically effective amount means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit risk ratio applicable to any treatment.
  • a pharmaceutical effect refers to a local or systemic effect in animals, particularly mammals, and more particularly humans, caused by a pharmacolog cal!' active substance or substances.
  • the term thus means any substance intended for use in diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and conditions in an animal or human.
  • therapeutically effective amount means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit risk ratio applicable to any treatment.
  • a pharmaceutically effective amount means that amount of such
  • metadoxine or metadoxine derivatives formulations of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to a selected treatment, as may be determined by the skilled artisan.
  • the term "effective amount” refers to the amount of a therapeutic reagent that when administered to a subject in an appropriate dose and regimen produces at least one desired result.
  • a "subject” or “patient” to be treated by a method of the invention may mean either a human or non-human animal, preferably a mammal.
  • the term "subject” as used herein may refer to a healthy individual, or a subject suffering Fragile X Syndrome or Autism Spectrum Disorder. .
  • the terms "subject” and “healthy subject” and “subject in need” and “patient in need” as used herein exclude subjects under alcohol influence following alcohol consumption of any form, alcoholics (alcohol addicts), and abstinent alcoholics.
  • salt adducf is meant to encompass a salt product of a direct addition of two or more distinct ions, wherein the overall charge of the salt adduct is zero.
  • the salt adduct comprises one positively charged moiety having a single positive charge functional group (i.e., the positively charged moiety is charged with H- 1 net charge) and one negatively charged moiety having a single negative charge functional group (i.e., the negatively charged moiety is charged with -1 net charge).
  • the salt adduct comprises one positively charged moiety having two positively charged functional groups, which may be the same or different (i.e., the positively charged moiety is charged with +2 net charge) and two negative!' charged moieties, which may be the same or different, and each having a single negative charged functional group (i.e., each negatively charged moiety is charged with -1 net charge).
  • the salt adduct comprises two positively charged moieties, which may be the same or different, having each one positively charged functional group (i.e., each positively charged moiety is charged with +1 net charge) and one negatively charged moiety, having two negatively charged functional groups, being the same or different (i.e., the negatively charged moiety is charged with -2 net charge).
  • the salt adduct comprises a positively charged moiety charged with +n net charge (originating from one or more positively charged functional groups, which may be the same or different), and a negatively charge moiety having -n (originating from one or more negatively charged functional groups, which may be the same or different) net charge, wherein n is an integer which may be equal to 1 , 2, 3, 4, 5 or 6.
  • a "positively charged moiety of a salt adduct" of the invention is the corresponding acid of pyridoxine, or any derivative thereof
  • the positive charge of the positively charged moiety stems from the protonated basic nitrogen atom of pyridoxine (as for example in compound (2)) or any derivative thereof (such as for example compounds of formula (I)).
  • the positively charged pyridoxine derivative is substituted with a positively charged functional group such as for example ⁇ NH 3 " , -CH 2 NH 3 , H 2 " , - HR. 2 + (wherein each R is independently a alkyl), which may, in some
  • embodiments be present in addition to the positively charged protonated basic aromatic nitrogen atom in the pyridine ring.
  • moieties of a salt adduct of the invention may contain each at least one chiral center, and thus may exist in, and be isolated as, any stereoisomer thereof including, enantiomers, diastereomers or any mixtures thereod including, but not limited to racemic mixtures.
  • the present invention includes any possible stereoisomer (e.g. enantiomers, diastereomers), any mixtures thereof including, but not limited to, racemic mixtures, of any of the individual moieties of a salt adduct of the invention.
  • each of the moieties of a salt adduct of the invention may be separated by conventional techniques, such as preparative chromatography.
  • the moieties of a salt adduct of the invention may be each prepared in any mixture of possible stereoisomers thereof, including but not limited to racemic mixtures thereof, or individual stereoisomers (e.g. enantiomers, diastereomers) may be prepared either by
  • bio-available means that at least some amount of a particular compound is present in the systemic circulation.
  • Formal calculations of oral bioavailability are described in terms of an F value ("Fundamentals of Clinical Pharmacokinetics," John G . Wegner, Drug Intelligence Publications; Hamilton, 111. 1975).
  • F values are derived from the ratio of the concentration of the parent drug in the systemic circulation (e.g., plasma) following intravenous administration to the concentration of the parent drug in the systemic circulation after
  • oral bioavailability within the scope of the present invention contemplates the ratio or F value of the amount of parent drug detectable in the plasma after oral administration compared to intravenous administration.
  • treating refers to mitigating, improving, relieving or alleviating at least on symptom of a condition, disease or disorder in a mammal, such as a human, or the improvement of an ascertainable measurement associated with a condition, disease or disorder. Treatment as used herein also encompasses treatment of healthy individuals.
  • metadoxine or metadoxine derivatives refers to any salt, conjugate, ester, complex or other chemical derivative of metadoxine or any of the moieties comprising the same, which, upon administration to a subject, is capable of providing (directly or indirectly) metadoxine or a metabolite or functional residue thereof, or measurable metadoxine activity.
  • physiologically compatible metadoxine derivative may be used interchangeably herein with the term “acceptable derivative” and refers to a functional, active, pharmaceutically acceptable derivative of metadoxine.
  • excipie t refers to an inactive substance used as a carrier for the active ingredient in a formulation.
  • controlled release refers to any formulation which delivers an agent at a control led rate for an extended time and is designed to achieve a desired agent level profile
  • sustained release is used in its conventional sense to refer to a formulation that provides for gradual release of an acti ve material over an extended period of time, which in certain embodiments may also further result in substantially constant blood levels over an extended time period, i.e., controlled release.
  • immediate release is used in its conventional sense to refer to a formulation that provides for non delayed or control led release of an active material upon administration .
  • half-life of a substance is the time it takes for a substance to lose half of its pharmacologic, physiologic, or other activity.
  • Biological half-life is an important pharmacokinetic parameter and is usually denoted by the abbreviation tin..
  • non-in vasive refers to modes of treatment which do not puncture the skin.
  • non-chronic administration may be used interchangeably herein with the term “acute administration” and refers to giving a measured or non-measured quantityor portion of a medication to a subject on a non-regular basis.
  • Non-chronic administration may be a single dose treatment or a multiple dose treatment, and may optionally be given over time. Typically but not always, a non-chronic administration is given to treat or prevent a non-chronic condition. Certain chronic conditions may also benefit from non-chronic administration of a metadoxine or metadoxine derivatives composition described herein.
  • chronic administration refers to giving a measured quantity of a medication on a regular basis to a subject.
  • chronic administration is to treat or prevent one or more chronic conditions, problems or diseases.
  • Chronic diseases have one or more of the following characteristics: they are permanent, leave residual disability, are caused by
  • nonreversible pathological alteration require special training of the patient for rehabilitation, or may be expected to require a long period of supervision, observation, or care.
  • single dose treatment refers to giving a measured quantity of a medication to be taken at one time. It is given to treat non-chronic conditions on an irregular basis, depending on personal need.
  • t max refers to the time to peak concentration. Cal culation of time at which maximum concentration occurs after a single dose administration is performed according to the formula:
  • Fmrl knockout mice ( 02) mice (The Dutch-Beigium Fragile X Consortium, 1994), initially obtained from the Jackson Laboratory, and wild type (WT) littermates were generated on a C57BL/6J background and repeatedly backcrossed onto a C57BL/6J background for more than eight generations.
  • the Fmrl knockout mice were housed in groups of the same genotype in a temperature and humidity controlled room with a ! 2 ⁇ h light-dark cycle (lights on from 7 am to 7 pm; testing was conducted during light phase). Room temperature and humidity were recorded continuously in the holding room while food and water were available ad libitum.
  • mice were housed in commercial plastic cages and experiments were conducted in line with the requirements of the UK Animals (Scientific Procedures) Act, 1986. All experiments were conducted with experimenters blind to genotype and drug treatment. Animals were allowed a minimum acclimatization period of one week prior to performing any experiment. No prophylactic or therapeutic treatment was administered during the acclimatization period.
  • mice were placed in a test arena/cage of the same order of magnitude in size as the adult's home cage (40 x 23 x 12 cm cage, with a Perspex lid to facilitate viewing the mice) with fresh wood chippings on the floor.
  • a background mouse odor was created by putting in some non-experimental mice into the apparatus prior to testing. Mice were transferred to the experimental room 10-15 min prior to testing.
  • a test subject and a juvenile were placed simultaneously into the test cage. The total duration and number of bouts of social investigation, defined as sniffing and close following ( ⁇ 2 cm from the tail) of the stimulus juvenile by the tested mouse, was assessed for 3 min. 30 min later, the test was repeated using the same stimulus juvenile.
  • Y-Maze Alternation Two tasks were implemented. The first was an unlearned assessment of spontaneous alternation between arm entries. The second was a spatial reference memory task in which the animal had to learn which of the two arms was baited with a food reward. The day prior to the start of the training, mice were allowed to freely explore the maze for 5 min. Next, they received two trials, one in which the food was located on the left arm and one in which the food was positioned on the right arm. This procedure prevented the development of a preference for one of the arms.
  • Y-Maze Water Maze A clear Perspex Y-maze was filled with 2 cm of water at 20°C. This motivated the mouse to leave the maze after paddling to an exit tube at the distal end of one arm. The maze was placed in the middle of a room surrounded by prominent visual cues.
  • the apparatus consisted of four successive, linearly arranged, increasingly anxiogenic alleys (each succeeding alley was painted a lighter color, had lower walls and/or was narrower than the previous alley) made of painted wood. Each section or alley was 25 cm long. Alley 1 had 25 cm high wal ls, was 8,5 cm wide, and was painted black, A 0,5 cm step down led to alley 2, which was again 8.5 cm wide, but had 1.3 cm high walls and was grey. A 1.0 cm step down led to al ley 3, which was 3,5 cm wide, had 0.8 cm high walls, and was white. A 0.4 cm step led down to alley 4, which was also white, but had 1.2 cm wide and 0.2 cm high walls.
  • the apparatus was elevated by anchoring the back of alley 1 to a stand, 50 cm high . Padding was provided under arms 3 and 4 in case a mouse fell off. Each mouse was placed at the closed end of alley 1 facing the wall. Timers were started 1) for the overall length of the test (5 min) + the latency to enter each arm, and 2) for the time spent in alley 1. When the mouse placed all 4 feet on to the next alley, it was considered to have entered the alley. Total time spent in each alley (ail four feet) was recorded.
  • Contextual Fear Conditioning In the fear conditioning experiment, mice were placed into a novel environment (dark chamber) and received pairings of a cue and electric footshock (0.2 mA for 1 sec (Study 1) or 0.7 mA for 0.5 sec (Study 2)). Subsequently, when tested in the original training context, mice displayed a natural defensive response termed freezing (Bianchard, 1969) or contextual fear conditioning. Freezing time was defined as the time that the mice spent in immobile behavior, except for respiration. The data was expressed as the percentage of the test period. 24 hours after a training session, mice were tested for 5 min in the training chamber with no shock presentation and observed for freezing behavior,
  • Phosphorylated ERK and Akt The Ras-Mek-ERK and PI3K-Akt-mToR signaling pathways are involved in mediating activity dependent alterations in gene transcription underlying changes in synaptic plasticity (Klami and Dever, 2004). Phosphorylated ERK and Akt protein expression was measured by western blot analysis as previously described by Lopez Verrilli (Lopez Verrilli et aL, 2009), The antibodies employed were a ti-phosphospecific antibodies against Akt ( 1/1000) and kinase (ERK) 1/2 (1/2000) (Cell Signaling Technology, Danvers, MA, LISA), The antibody against phospho-ERK detects phosphorylation at phospho-ER l/2
  • Akt and ERK 1/2 protein content and phosphorylated ERK and Akt were evaluated by blotting membranes with antiphospho-Akt (1/1000) and anti phospho-ERK antibodies (1/2000) (Cell Signaling Technology, Danvers, MA, USA). Akt or ERK phosphorylation was normalized to protein content in the same sample and expressed as % of change with respect to basal conditions, considering basal levels as 100%. Protein loading was evaluated by stripping and re-blotting membranes with ⁇ -actin antibody (1/1000) (Sigma-Aldrich, St. Louis, MO, USA).
  • Phosphory lated ERK and Akt protein expression in blood lymphocytes was measured by flow cytometry.
  • a FACStar plus Becton Dickinson
  • GST green fluorescence from FJTC
  • the mean FITC fluorescence Intensity was calculated in relation to the fluorescence of reference cells.
  • the mea cellular fluorescence intensity (MFI) is directly proportional to the mean number of Ab molecules bound per cell.
  • Filopodia density of GFP transfected neurons was quantified by performing Sholl analyses of stacked Zeiss confocal generated images (40 x objective, stack of 20 ⁇ 0.2 ⁇ ). With Metamorph software, concentric equally spaced circles (every 20um) were drawn around the cell soma of each neuron and subsequently, the amount of filopodia was counted per circle. Averages of counts were compared with unpaired two-tai led Student's T-tests.
  • width was defined as the maximum distance peipendicular to the long axis of the spine. Measurements were compared with unpaired two tailed Student's T-tests and ANOVA corrected for multiple comparisons.
  • EXAMPLE 2 THE EFFECT OF METADOXINE (100 to 200 mg/kg) TREATMENT ON LEARNING AND MEMORY DEFICITS AND BIOCHEMICAL ABNORMALITIES ⁇ THE Fmrl KNOCKOUT MOUSE MODEL OF FRAGILE X SYNDROME (Study 1)
  • Y-maze Reference Memory Task The effect of seven days of once daily treatment with vehicle or 150 mg/kg metadoxine on rewarded reference memory learning in groups of N ::: 10 WT or Fmrl knockout mice is shown in Figure 3, Panel B.
  • Vehicle-treated Fmrl knockout mice made less appropriate arm entries than vehicle-treated WT mice (pO.0001).
  • Metadoxine treatment reduced this deficit (p ⁇ 0.0001) compared to vehicle-treated Fmrl knockout mice, such that metadoxine-treated Fmrl knockout mice did not differ from metadoxine-treated WT mice, Metadoxine treatment had no effect on WT mice.
  • Vehicle-treated Fmrl knockout mice showed a greater latency to reach the correct arm compared to vehicle-treated WT mice (p ⁇ 0.0001).
  • Metadoxine treatment reduced this deficit compared to vehicle treatment in / ⁇ ;;>/ ⁇ / knockout mice (p ⁇ 0.0001), this reversal being partial since metadoxine-treated Fmrl knockout mice responded more slowly than WT mice (p ⁇ 0.00Gl ).
  • the successive alleys test effectively measured anxiety (latency to enter the Alley 1 ) and hyperactivity (Alleys 2 to 4). Progression from Alley 1 through the successive Alleys 2, 3, and 4 was associated with exposure to an increasingly brightly colored environment with increasingly lower wails and narrower, more exposed open arms. Time spent on, and entries into, the open arms indicated anxiety; conversely, increasing time spent in more open arms reflected hyperactivity. These factors allowed for a sensitive test bracketing a range of anxiety-like behaviors together with hyperactivity.
  • Alley 1 The Fmrl knockout mice showed more anxiety than WT mice (p ⁇ Q.001). Fmrl knockout mice treated with metadoxine showed an amelioration in anxiety compared with the vehicle treated Fmrl knockout mice (p ⁇ 0.001), such that complete normalization occurred. There was no difference between the metadoxine-treated Fmrl knockout and metadoxine-treated WT mice. Also, metadoxine treatment had no effect on WT mice.
  • Alley 2 WT mice showed less activity in Alley 2 when compared with the Fmrl knockout mice (p ⁇ 0.0001). Treatment with metadoxine reduced hyperactivity in the Fmrl knockout mice (p ⁇ 0.001 ), although this reversal of hyperactivity was partial since metadoxine- treated Fmrl knockout and WT mice differed (p ⁇ 0.001). Metadoxme treatment had no effect on WT mice.
  • Alley 3 Fmrl knockout mice showed hyperactivity compared to WT mice (p ⁇ 0.0001). This hyperactivity was not reversed by metadoxine, since metadoxine-treated Fmrl knockout mice did not differ from vehicle-treated Fmrl knockout mice. Metadoxme treatment had no effect on WT mice.
  • Vehicle-treated Fmrl knockout mice (KO-V) showed a deficit in learning in the contextual fear conditioning paradigm when compared with vehicle-treated WT mice ( WT-V) as reflected in a reduction in freezing during the test session (Fig. 7 (p ⁇ 0.QQ01)).
  • Metadoxme administration reversed the learning deficit effect in Fmrl knockout mice (p ⁇ 0.0001 KO-M-150 vs. KO-V). This was a complete reversal such that metadoxme-treated KO mice did not differ from metadoxine-treated WT mice.
  • Fmrl knockout mice showed an increased number of sniffing bouts (p ⁇ 0.0001 ) (See Fig. 8, Panel A) and a reduced duration of sniffing (p ⁇ 0.0001 ) (See Fig. 8, Panel C) compared to WT mice. These social interaction deficits are consistent with those reported by other researchers in Fmrl knockout mice (Thomas et al., 201 1). For both number of bouts and duration of sniffing, treatment with metadoxme produced reversals of abnormalities in Fmrl knockout mice (pO.0001 KO-M-150 vs.
  • Figure 10 shows the effect of administration of once daily metadoxine at doses of 150 mg/kg ip or 150 and 300 mg/kg orally for seven days on contextual fear conditioning in two month old Fmrl knockout and WT mice.
  • Panel A shows contextual fear conditioning data from Fmrl knockout and WT mice after ip and oral treatment with vehicle. There were no differences related to the route of administration of vehicle. Fmrl knockout mice showed a reduction in freezing behavior compared to WT mice after vehicle treatment via ip and oral routes (p ⁇ 0.0001 in each case).
  • Panel B shows the effect of metadoxine treatment via both routes of administration in WT mice. No effects were seen.
  • Panel C shows that ip 150 mg/kg and oral 150 and 300 mg/kg metadoxine treatment in Fmrl knockout mice reversed the decrease in freezing behavior seen in Fmrl knockout mice (p ⁇ G.01, p ⁇ 0.0001, and j 0.000 i . for KO-M-ip, KO-M-po 150, and KO-M-po 300 vs. KO-V-ip and KO-V o, respectively).
  • the effect of administration with 150 mg po metadoxme did not differ from the effect of administration of 300 mg/kg po metadoxine.
  • the effect of 150 and 300 mg/kg oral metadoxme in Fmrl knockout mice did not differ from the effect of 150 mg/kg ip metadoxme. In each case, the reversal was complete since metadoxine-treated Fmrl knockout mice did not differ from metadoxine-treated WT mice.
  • Figure 11 shows the effect of administration of once daily metadoxine at doses of 150 mg/kg ip or 150 and 300 mg/kg orally for seven days on social approach and social memory in Fmrl knockout and WT mice.
  • Figure 1 1 Panel A shows the effect of vehicle or metadoxine at 150 mg/kg ip or 150 and 300 mg/kg orally on social approach behavior in Fmrl knockout or WT mice. After ip or oral treatment with vehicle, the duration of sniffing behavior in Fmrl knockout mice was reduced compared to WT mice (p ⁇ 0.0001 for each). Metadoxine treatment at any dose was without effect on WT mice.
  • metadoxine treatment at 150 mg/kg ip, 150 mg kg, and 300 mg/kg orally produced reversals of the social approach deficit seen in Fmrl knockout mice (p ⁇ 0.0001 for KO-M-po 150 and KO-M-po300 vs. KO-V po,
  • Peripheral Lymphocytes Figure 12 shows the effect of administration of once daily metadoxine at doses of 150 mg/kg ip or 150 mg/kg and 300 mg/kg orally for 7 days on
  • lymphocyte pAkt Fig. 12, Panel A
  • pERK Fig. 12, Panel B
  • FIG. 12 Panel A
  • vehicle-treated Fmrl knockout mice exhibited increased phosphorylation of lymphocyte Akt (pO.0001 for both ip and oral administration) compared to WT mice receiving equivalent vehicle treatment.
  • Panel B shows that vehicle-treated Fmrl knockout mice showed increased phosphorylation of lymphocyte ERK (p ⁇ Q.00Ql for both ip and oral administration) compared to WT mice receiving equivalent vehicle treatment, Treatment with once daily metadoxine at 150 mg/kg ip or oral doses of 150 mg/kg or 300 mg/kg for 7 days normalized overactivated ERK such that pERK levels did not differ between metadoxine-treated Fmrl knockout mice and WT mice receiving the same treatment.
  • Figure 13 shows the effect of administration of 150 mg/kg metadoxine for seven days on pERK levels in hippocampus, pre-frontal cortex, and striatum.
  • pERK levels were increased in Fmrl knockout mice compared to WT mice in all three brain regions (p ⁇ 0.0001 in all cases).
  • pERK levels were decreased in metadoxine-treated Fmrl knockout mice compared to vehicle-treated Fmrl knockout mice (p ⁇ 0.0001 in all cases).
  • the effect in pre-frontal cortex was partial, the KG-V and KO-M groups remained different (p ⁇ 0.05). Metadoxine was without effect on WT mice.
  • Figure 14 shows the effect of administration of 150 mg/kg metadoxme for seven days on Akt levels in hippocampus, pre-frontal cortex and striatum.
  • Akt levels were increased in Fmrl knockout mice compared to WT mice in all three brain regions (p ⁇ 0.0001 in all cases).
  • pAkt levels were decreased in metadoxine-treated Fmrl knockout mice compared to vehicle- treated Fmrl knockout mice in all three brain regions (p ⁇ 0.0001 in all cases).
  • Metadoxine was without effect on WT mice. Reduction in brain and blood elevated levels of phosphorylated ERK and Akt correlated with the improved behavioral outcomes of Fmrl knockout mice, suggesting that the phosphorylation levels are hiomarkers of metadoxme treatment response
  • Figure 15 shows the effect of treatment for five hours with 300 ⁇ metadoxme. Dendrites were divided into 10 segments of 10 ⁇ , each based on distance from the soma (proximal to distal, left to right). Spine density was increased in neurons from Fmrl
  • FIG. 15 Panel A shows the density of neuronal filopodia.
  • Primary hippocampal neurons from Fmrl knockout mice displayed an increased density of filopodia (p ⁇ 0.001).
  • Treatment with 300 ⁇ metadoxine reduced the aberrant increase in density of neuronal filopodia in Fmrl knockout mice (p ⁇ 0.001).
  • Neurons from Fmrl knockout mice showed filopodia with characteristics of immaturity, being longer (Fig. 15, Panel B (p ⁇ 0.01)) and narrower (Fig. 15, Panel C (p ⁇ 0.01)).
  • Treatment with metadoxine reversed this increase in filopodia length ( Fig. 1 5, Panel B (p ⁇ 0.01)) and reversed the decrease in width (Fig. 15, Panel C (pO.OOl)).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Urology & Nephrology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Medicines Containing Plant Substances (AREA)
EP14830858.8A 2013-09-09 2014-09-09 Methods of treating fragile x syndrome and related disorders Withdrawn EP3043792A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361875384P 2013-09-09 2013-09-09
US14/038,258 US20150073023A1 (en) 2013-09-09 2013-09-26 Method Of Treating Fragile X Syndrome And Related Disorders
US201461991351P 2014-05-09 2014-05-09
PCT/IB2014/002398 WO2015033224A2 (en) 2013-09-09 2014-09-09 Methods of treating fragile x syndrome and related disorders

Publications (1)

Publication Number Publication Date
EP3043792A2 true EP3043792A2 (en) 2016-07-20

Family

ID=52629036

Family Applications (2)

Application Number Title Priority Date Filing Date
EP14776939.2A Withdrawn EP3044589A1 (en) 2013-09-09 2014-09-09 Methods of determining response to therapy
EP14830858.8A Withdrawn EP3043792A2 (en) 2013-09-09 2014-09-09 Methods of treating fragile x syndrome and related disorders

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP14776939.2A Withdrawn EP3044589A1 (en) 2013-09-09 2014-09-09 Methods of determining response to therapy

Country Status (12)

Country Link
EP (2) EP3044589A1 (ru)
JP (2) JP2016530291A (ru)
KR (2) KR20160086818A (ru)
CN (2) CN105917225A (ru)
AU (2) AU2014315026A1 (ru)
CA (2) CA2922901A1 (ru)
EA (2) EA201690559A1 (ru)
IL (2) IL244343A0 (ru)
MX (2) MX2016003006A (ru)
SG (2) SG11201601830PA (ru)
TW (2) TW201605443A (ru)
WO (2) WO2015033224A2 (ru)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105431442B (zh) * 2013-07-31 2018-08-07 Udc 爱尔兰有限责任公司 发光的二氮杂苯并咪唑碳烯金属配合物
US20150073023A1 (en) 2013-09-09 2015-03-12 Alcobra Ltd. Method Of Treating Fragile X Syndrome And Related Disorders
CN108538365A (zh) * 2017-03-24 2018-09-14 华东师范大学 一种基于数据分析技术的自闭症社交能力评估系统
EP3773538A1 (en) * 2018-04-13 2021-02-17 Healx Limited Treatment of fragile x syndrome
KR20190121569A (ko) * 2018-04-18 2019-10-28 건국대학교 글로컬산학협력단 아그마틴 및 이의 약학적으로 허용 가능한 염을 포함하는 취약 x 증후군 예방 또는 치료용 약학조성물
CA3102786A1 (en) 2018-06-07 2019-12-12 Ovid Therapeutics Inc. Use of (s)-3-amino-4-(difluoromethylenyl) cyclopent-1-ene-1-carboxylic acid and related compounds, (1s,3s)-3-amino-4-(difluoromethylidene) cyclopentane-1-carboxylic acid and vigab atrin in the treatment of developmental disorders
US20230024384A1 (en) * 2020-01-08 2023-01-26 Neuroventi Composition comprising lisuride compound for treating fragile x syndrome or related developmental disorders
KR102398219B1 (ko) * 2020-02-07 2022-05-17 주식회사 뉴로벤티 릴메니딘 화합물을 유효성분으로 포함하는 취약 x 증후군 또는 관련 발달 장애 치료용 조성물

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1131856B (it) 1980-06-30 1986-06-25 Baldacci Lab Spa Composizione farmaceutica per il trattamento di intossicazioni alcooliche
US20020155170A1 (en) * 2000-11-30 2002-10-24 Walsh William John Nutrient supplements and methods for treating autism and for preventing the onset of autism
IL187159A0 (en) 2007-07-03 2009-02-11 Gur Megiddo Use of metadoxine in relief of alcohol intoxication
EA023758B1 (ru) * 2009-06-25 2016-07-29 Алкобра Лтд. Применение метадоксина для лечения синдрома дефицита внимания/гиперактивности (adhd/add)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2015033224A2 *

Also Published As

Publication number Publication date
CA2923421A1 (en) 2015-03-12
IL244453A0 (en) 2016-04-21
KR20160078956A (ko) 2016-07-05
JP2016530536A (ja) 2016-09-29
SG11201601830PA (en) 2016-04-28
TW201605443A (zh) 2016-02-16
CN105517546A (zh) 2016-04-20
AU2014315026A1 (en) 2016-03-24
SG11201601605YA (en) 2016-04-28
EA201690557A1 (ru) 2016-07-29
JP2016530291A (ja) 2016-09-29
MX2016003002A (es) 2016-09-08
AU2014316779A1 (en) 2016-03-17
MX2016003006A (es) 2016-06-10
CN105917225A (zh) 2016-08-31
WO2015035402A1 (en) 2015-03-12
WO2015033224A2 (en) 2015-03-12
WO2015033224A3 (en) 2015-07-02
CA2922901A1 (en) 2015-03-12
TW201606304A (zh) 2016-02-16
IL244343A0 (en) 2016-04-21
EP3044589A1 (en) 2016-07-20
KR20160086818A (ko) 2016-07-20
EA201690559A1 (ru) 2016-08-31

Similar Documents

Publication Publication Date Title
WO2015033224A2 (en) Methods of treating fragile x syndrome and related disorders
KR102014883B1 (ko) 근위축성 측삭 경화증 치료용 신규 조성물
JP5875191B2 (ja) Cmt及び関連障害を処置するための新たな組成物
CN104127434B (zh) 使用唑尼沙胺和阿坎酸治疗阿尔茨海默病和相关病症的组合组合物
EP1793671B1 (en) Use of memantine (namenda) to treat autism, compulsivity, and impulsivity
US9808446B2 (en) Method for decreasing symptoms of alcohol consumption
EA024465B1 (ru) ПРИМЕНЕНИЕ ЛЕВОСИМЕНДАНА ИЛИ ЕГО СОЛЕЙ ДЛЯ ПРИГОТОВЛЕНИЯ МЕДИКАМЕНТА ДЛЯ ЛЕЧЕНИЯ БОЛЕЗНИ АЛЬЦГЕЙМЕРА И РОДСТВЕННЫХ РАССТРОЙСТВ ИЛИ ДЛЯ ЗАЩИТЫ ЭНДОТЕЛИАЛЬНЫХ И/ИЛИ НЕРВНЫХ КЛЕТОК ОТ Аβ ТОКСИЧНОСТИ И СПОСОБ ЛЕЧЕНИЯ УКАЗАННЫХ ЗАБОЛЕВАНИЙ
JP5677946B2 (ja) シャルコー・マリー・トゥース病および関連した障害を処置するためのピロカルピンおよびメチマゾールの組み合わせ
JP2007517040A (ja) 睡眠の質を改善するためのメラトニン併用療法
CZ20023121A3 (cs) Farmaceutické prostředky pro léčení diabetických komplikací a neuropatie a jejich použití
JP2010504338A (ja) 筋萎縮性側索硬化症(ALS)の治療のための水素化ピリド[4,3−b]インドール
US20050215521A1 (en) Modafinil combination therapy for improving sleep quality
EP3388062A1 (en) Compositions for treating parkinson's disease
EP3223815B1 (en) Combination of ibudilast and riluzole and methods of using same
TW202112359A (zh) sGC活化劑於治療眼科疾病之用途
RU2336870C2 (ru) Применение l-бутилфталида для изготовления лекарственного средства для профилактики и лечения церебрального инфаркта
US9851354B2 (en) Methods of treating fragile X syndrome and related disorders
UA109540C2 (uk) Сполуки для пригнічення розладу периферійних нервів, викликаного протираковим агентом
US20210161886A1 (en) Methods of treating depressive disorders
CN101541323B (zh) 吲唑甲氧基烷酸用于降低甘油三酯、胆固醇和葡萄糖水平的用途
IL303006A (en) Use of pyridofidine and analogues for the treatment of Rett syndrome
CN114072154A (zh) 用于治疗精神障碍的加波沙朵和锂的组合物
JP2017517574A (ja) 神経発生性疾患および障害を処置する方法
US20230165852A1 (en) Method for treating central nervous system disorders using dopamine d3 partial agonists
CN117479931A (zh) 用于治疗孤独症谱系障碍(asd)的方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20160324

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: MEGIDDO, DALIA

Inventor name: DANIELY, YARON

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1226314

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20180404

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1226314

Country of ref document: HK