US20200289553A1 - Compositions and Methods for the Treatment of Metabolic Conditions - Google Patents

Compositions and Methods for the Treatment of Metabolic Conditions Download PDF

Info

Publication number
US20200289553A1
US20200289553A1 US16/770,807 US201816770807A US2020289553A1 US 20200289553 A1 US20200289553 A1 US 20200289553A1 US 201816770807 A US201816770807 A US 201816770807A US 2020289553 A1 US2020289553 A1 US 2020289553A1
Authority
US
United States
Prior art keywords
pharmaceutical grade
subject
acid
buffer solution
composition
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.)
Abandoned
Application number
US16/770,807
Other languages
English (en)
Inventor
James Ervin
Hendrik J. Van Wyk
Brian D. Denomme
Mariette L. Van Wyk
Peter Pacult
Michael A. Volk
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.)
Reven IP Holdco LLC
Original Assignee
Reven Pharmaceuticals Inc
Reven IP Holdco LLC
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 Reven Pharmaceuticals Inc, Reven IP Holdco LLC filed Critical Reven Pharmaceuticals Inc
Priority to US16/770,807 priority Critical patent/US20200289553A1/en
Publication of US20200289553A1 publication Critical patent/US20200289553A1/en
Assigned to REVEN IP HOLDCO LLC reassignment REVEN IP HOLDCO LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LANGE, PETER B.
Assigned to REVEN IP HOLDCO LLC reassignment REVEN IP HOLDCO LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REVEN PHARMACEUTICALS, INC.
Assigned to REVEN IP HOLDCO LLC reassignment REVEN IP HOLDCO LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REVEN, LLC
Assigned to REVEN, LLC reassignment REVEN, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DENOMME, Brian David, PACULT, Peter, VAN WYK, HENDRIK JOHANNES PETRUS, VAN WYK, Mariette Luise
Assigned to REVEN PHARMACEUTICALS, INC. reassignment REVEN PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VOLK, MICHAEL A., DENOMME, Brian David, PACULT, Peter, VAN WYK, HENDRIK JOHANNES PETRUS, VAN WYK, Mariette Luise, ERVIN, JAMES
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/375Ascorbic acid, i.e. vitamin C; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic 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
    • A61K31/435Heterocyclic 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
    • A61K31/455Nicotinic acids, e.g. niacin; Derivatives thereof, e.g. esters, amides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • A61K31/51Thiamines, e.g. vitamin B1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/525Isoalloxazines, e.g. riboflavins, vitamin B2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • 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
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • A61K33/10Carbonates; Bicarbonates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/14Alkali metal chlorides; Alkaline earth metal chlorides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/20Elemental chlorine; Inorganic compounds releasing chlorine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0004Homeopathy; Vitalisation; Resonance; Dynamisation, e.g. esoteric applications; Oxygenation of blood
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • 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
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • 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
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • A61K9/0031Rectum, anus
    • 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
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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

Definitions

  • the present invention relates to stable therapeutic compositions comprising pharmaceutical grade acids and pH buffering agents.
  • the present invention also is directed to methods of treatment for conditions and disorders characterized by mitochondrial dysfunction, metabolic conditions, diabetic conditions, cardiovascular conditions, and bone and tissue modeling dysfunction, comprising administration of compositions of the present disclosure.
  • Acid-base homeostasis is the ability of an organism to maintain a condition of equilibrium or stability within its internal environment, particularly when faced with external changes.
  • Some examples of homeostatically-controlled systems in humans include the regulation of a constant body temperature, blood glucose levels, and extracellular ionic species concentrations.
  • Acid-base homeostasis relates to the proper balance of acids and bases in extracellular fluids, i.e., the pH of the extracellular fluid. In humans, the pH of plasma is approximately 7.4, and is tightly maintained around that value by three interconnected control systems: 1) buffering agents, including bicarbonate, phosphate, and proteins, 2) the respiratory system, which impacts the partial pressure of carbon dioxide in blood plasma, and 3) the renal system, which excretes waste acids and bases.
  • Acid homeostasis is also influenced by metabolic load, which serves as a primary source of acid in the body. For instance, a high glucose diet can increase total acid burden from metabolic sources, to place a bigger burden on acid homeostasis control mechanisms.
  • Cardiovascular diseases may also be caused by a poor diet and sedentary lifestyle, and include coronary heart disease (heart attacks), cerebrovascular disease, raised blood pressure (hypertension), peripheral artery disease, rheumatic heart disease, congenital heart disease and heart failure.
  • coronary heart disease heart attacks
  • cerebrovascular disease cerebrovascular disease
  • raised blood pressure hypertension
  • peripheral artery disease peripheral artery disease
  • rheumatic heart disease congenital heart disease and heart failure.
  • Such dysfunctional conditions of the heart, arteries, and veins impair the supply of oxygen to vital life-sustaining organs, including the brain and the heart itself.
  • Heart attacks and strokes are mainly caused by a blockage in the inner walls of the blood vessels that prevents blood from flowing to the heart or the brain.
  • Arteriosclerosis also called atherosclerosis
  • the present disclosure provides a stable therapeutic composition formulated for intravenous administration to a subject, comprising an intravenous buffer solution, comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the pharmaceutical grade acid is hydrochloric acid, ascorbic acid, acetic acid, (other physiologically acceptable acids), or a combination thereof.
  • the at least one pH buffering agent is sodium bicarbonate, a phosphate, organic acid, organic amine, ammonia, citrate buffer, a synthetic buffer creating specific alkaline conditions (e.g., tris-hydroxymethyl amino methane), (other physiologically acceptable buffers), or a combination thereof.
  • the composition further comprises one or more ingredients selected from the group consisting of vitamins, salts, acids, amino acids or salts thereof, and stabilized oxidative species.
  • the composition further comprises ascorbic acid.
  • the composition comprises dehydroascorbic acid
  • the composition comprises other recognized antioxidant defense compounds, including nonenzymatic compounds, such as tocopherol (aTCP), coenzyme Q10 (Q), cytochrome c (C) and glutathione (GSH), and enzymatic components including manganese superoxide dismutase (MnSOD), catalase (Cat), glutathione peroxidase (GPX), phospholipid hydroperoxide glutathione peroxidase (PGPX), glutathione reductase (GR); peroxiredoxins (PRX3/5), glutaredoxin (GRX2), thioredoxin (TRX2) and thioredoxin reductase (TRXR2).
  • nonenzymatic compounds such as tocopherol
  • the composition is formulated in hypotonic, isotonic, or hypertonic form. In some embodiments, the composition is formulated for intravenous, bolus, dermal, oral, otic, suppository, buccal, ocular, or inhalation delivery. In some embodiments, the composition is formulated as a topical liquid, gel, or paste. In some embodiments, the composition is formulated for ocular administration in the form of eye drops. In some embodiments, the composition is lyophilized or frozen. In some embodiments, the composition is stored in a spectral-blocking vial. In some embodiments, the composition is formed by combining components from two or more vials.
  • the present disclosure provides a stable therapeutic composition formulated for intravenous administration to a subject comprising pharmaceutical grade 900 ⁇ 90 mg of L-Ascorbic Acid; 63.33 ⁇ 6.33 mg Thiamine HCl; 808 ⁇ 80.8 mg of Magnesium Sulfate; 1.93 ⁇ 0.193 mg of Cyanocobalamin; 119 ⁇ 11.9 mg of Niacinamide; 119 ⁇ 11.9 mg of Pyridoxine HCl; 2.53 ⁇ 0.253 mg of Riboflavin 5′Phosphate; 2.93 ⁇ 0.293 mg of Calcium D-Pantothenate; 840 ⁇ 84 mg of Sodium Bicarbonate; 4.5 ⁇ 0.45 mM of HCl; and water in an amount to obtain a final composition volume of 20 mL.
  • the composition further comprises 100 ⁇ 10 mg of dehydroascorbic acid.
  • the present disclosure provides a method of treating or ameliorating acidosis in a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of treating or ameliorating base excess in a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of elevating blood oxygen in a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the method comprises elevating the pO 2 in the venous blood in a subject.
  • the present disclosure provides a method of treating or ameliorating a mitochondrial disorder, metabolic disorder, a condition associated with diabetes or a cardiovascular dysfunction in a subject in need thereof, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the metabolic disorder is diabetes, insulin resistance, glucose intolerance, hyperglycemia, hyperinsulinemia, obesity, hyperlipidemia, or hyperlipoproteinemia.
  • the condition associated with diabetes is hypertension, hyperlipidemia, fatty liver disease, nephropathy, neuropathy, renal failure, retinopathy, diabetic ulcer, cataracts, insulin resistance syndromes and cachexia.
  • the cardiovascular dysfunction is coronary heart disease, cerebrovascular disease, hypertension, peripheral artery disease, occlusive arterial disease, angina, rheumatic heart disease, congenital heart disease, heart failure, cardiac insufficiency, palpitations, supraventricular tachycardia, fibrillation, faintness, dizziness, fatigue, migraine, high levels of total blood cholesterol and/or LDL cholesterol, low level of HDL cholesterol, high level of lipoprotein, infections of the heart such as carditis and endocarditis, diabetic ulcer, thrombophlebitis, Raynaud's disease, anorexia nervosa, claudication, gangrene, atherosclerosis and peripheral artery disease.
  • the mitochondrial disorder is a neurodegenerative disorder, a cardiovascular disease, a metabolic syndrome, an autoimmune disease, a neurobehavioral or psychiatric disease, a gastrointestinal disorder, a fatiguing illness, a chronic musculoskeletal disease, or a chronic infection.
  • the ocular condition is glaucoma, macular degeneration, eye floaters, ocular lens stiffening, or light sensitivity.
  • the composition further comprises dehydroascorbic acid. In some embodiments, the composition further comprises one or more of a magnesium ion source, a potassium ion source, and a calcium ion source. In some embodiments, the composition further comprises one or more of a B vitamin, vitamin C, and vitamin K.
  • the composition further comprises other recognized antioxidant defense compounds including nonenzymatic compounds such as tocopherol (aTCP), coenzyme Q10 (Q), cytochrome c (C) and glutathione (GSH), and enzymatic components including manganese superoxide dismutase (MnSOD), catalase (Cat), glutathione peroxidase (GPX), phospholipid hydroperoxide glutathione peroxidase (PGPX), glutathione reductase (GR); peroxiredoxins (PRX3/5), glutaredoxin (GRX2), thioredoxin (TRX2) and thioredoxin reductase (TRXR2).
  • nonenzymatic compounds such as tocopherol (aTCP), coenzyme Q10 (Q), cytochrome c (C) and glutathione (GSH)
  • enzymatic components including manganese superoxide dismutase (MnSOD), catalase (Cat), glut
  • the composition is formulated in hypotonic, isotonic, or hypertonic form.
  • the composition is administered intravenously, by bolus, dermally, orally, otically, via suppository, buccally, ocularly, or via inhalation.
  • the administering comprises introducing said composition by infusion over a period of about 1 minute to about 1 hour, and said infusion is repeated as necessary over a period of time selected from about 1 day to about 1 year.
  • the present disclosure provides a method of modifying the metabolism of a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of treating a central nervous system disorder in a subject in need thereof, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of treating chronic wounds of a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of enhancing mental or physical performance of a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of reducing lactate burden of a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the lactate burden is acidosis, sepsis, or multiple system atrophy (MSA).
  • the lactate burden is the result of physical exertion.
  • the present disclosure provides a method of improving hypoxic stress of a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the present disclosure provides a method of removing vascular plaque from the arteries of a subject, the method comprising administering to the subject a stable therapeutic composition comprising an intravenous buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, in a sterile aqueous solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7.
  • the methods of the invention provide a buffer solution that is sufficient to reduce the physiological bloodstream pH of a subject by 0.01 to 1.1.
  • the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.015 to 0.075.
  • the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.02 to 0.05.
  • the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.01 to 0.15.
  • the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.01 to 0.2.
  • the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.02 to 0.05.
  • the buffer solution has a buffer capacity sufficient to sustain the reduction of the physiological bloodstream pH of the subject for between 1 minute and 1 week. In other embodiments of the invention, the buffer solution has a buffer capacity sufficient to sustain the reduction of the physiological bloodstream pH of the subject for between 1 minute and 1 hour.
  • the subject is a human or veterinary subject.
  • the present disclosure provides a kit comprising (a) a first vial containing a stable therapeutic composition comprising a buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, wherein the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.1 to 1.1, and wherein the buffer solution has a buffer capacity sufficient to sustain the reduction of the physiological bloodstream pH of the subject for between 1 minute and 1 week; and (b) instructions for use.
  • kits comprising (a) a first vial containing an intravenous buffer solution comprising at least one pharmaceutical grade acid in a sterile aqueous solution;
  • a second vial containing at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution wherein, when combined, the contents of the two vials form an intravenous buffer solution, wherein the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject, and wherein the selection of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent is effective to provide a buffer solution pH of between 4 and 7.7; and (c) instructions for use.
  • FIG. 1 depicts a diagram of the typical chemiosmotic gradient of hydrogen ions between the inner-membrane and matrix in a normally functioning mitochondria in a mammalian cell.
  • FIG. 2 depicts a diagram of the reduced chemiosmotic gradient of hydrogen ions in a mitochondria in a mammalian cell with a dysfunctional metabolism, as may occur after a prolonged exposure to a poor diet, or lack of exercise.
  • FIG. 3 depicts a diagram of the chemiosmotic flow of ions into and out of the cell of a subject having a hypoxic crisis, or as observed in phases of acid-base disturbance, such as during or following exercise, or as observed during or following use of the composition of the invention.
  • FIG. 4 depicts a diagram of the chemiosmotic flow of ions into and out of the cell of a subject having had the hypoxic crisis corrected by use of the composition of the invention.
  • FIG. 5 shows a diagram of the amplitude and duration of an acid state shift caused by different formulations of compositions of the present disclosure.
  • FIGS. 6, 7, 8, 9, 10 and 11 show a graphic representation of the pH and HCO 3 ⁇ response
  • FIG. 6 Acid Shifting Composition; Dose 1, Day 1
  • sO 2 , pCO 2 , pO 2 response FIG. 7
  • FIG. 9 Acid Shifting Composition with Vitamins and Minerals; Dose 4, Day 6
  • FIG. 10 Acid Shifting Composition with Vitamins and Minerals; Dose 5, Day 8); and sO 2 , pCO 2 , pO 2 response ( FIG. 11 —Acid Shifting Composition with Vitamins and Minerals; Dose 5, Day 8) of Subject 2, after administration of the therapeutic composition.
  • FIGS. 12 and 13 show a graphic representation of the pH and HCO 3 ⁇ response ( FIG. 12 —Acid Shifting Composition; Dose 1, Day 8); and sO 2 , pCO 2 , pO 2 response ( FIG. 9 —Acid Shifting Composition; Dose 1, Day 8) of Subject 3, after administration of the therapeutic composition.
  • the term “mammal” refers to humans as well as all other mammalian animals.
  • the term “mammal” includes a “subject” or “patient” and refers to a warm-blooded animal. It is understood that guinea pigs, dogs, cats, rats, mice, horses, goats, cattle, sheep, zoo animals, livestock, primates, and humans are all examples of animals within the scope of the meaning of the term.
  • a mammal in need thereof may be a subject who could have been, but is not required to have been, diagnosed as suffering from the condition intended to be treated.
  • the present method is directed to conditions that are noticeable to the subject and the subject wishes to treat or ameliorate the condition without a formal diagnosis.
  • a mammal in need thereof is one who has been diagnosed as having a condition and is in need of specific treatment.
  • a mammal may also be functioning normally relative to common standards but electively seeks to enhance performance for various purposes, such as for enhanced mental acuity or athletic interests.
  • subject and “patient” are used interchangeably, and are meant to refer to any mammal, including humans, that has, or is at risk of developing, a dysfunctional cardiovascular condition.
  • the subject or patient is typically human, however, other suitable subjects or patients include, but are not limited to, laboratory animals, such as mouse, rat, rabbit, or guinea pig, farm animals and domestic animals or pets. Non-human primates are also included.
  • a “therapeutically effective amount” is an amount effective to elicit a cellular response that is clinically significant.
  • the terms “treating” and “ameliorating” are intended to refer to all processes wherein there may be a slowing, interrupting, arresting, or stopping of the progression of the condition or symptoms, and does not necessarily indicate a total elimination of the underlying condition.
  • the terms also encompass the administration of a pharmaceutical grade, physiological component, or natural physiological buffer composition wherein the mammal has a condition or symptom or a predisposition towards a condition or symptom, where the purpose is to cure, heal, alleviate, relieve, alter, improve or affect the condition or symptom or the predisposition to the same. Also contemplated is preventing the condition or symptom or the predisposition to the same, by prophylactically administering a pharmaceutical grade buffer composition as described herein.
  • a “pharmaceutical grade” means that certain specified biologically active and/or inactive components in the drug must be within certain specified absolute and/or relative concentration, purity and/or toxicity limits and/or that the components must exhibit certain activity levels, as measured by a given bioactivity assay.
  • a “pharmaceutical grade compound” includes any active or inactive drug, biologic or reagent, for which a chemical purity standard has been established by a recognized national or regional pharmacopeia (e.g., the U.S. Pharmacopeia (USP), British Pharmacopeia (BP), National Formulary (NF), European Pharmacopoeia (EP), Japanese Pharmacopeia (JP), etc.).
  • Pharmaceutical grade further incorporates suitability for administration by means including topical, ocular, parenteral, nasal, pulmonary tract, mucosal, vaginal, rectal, intravenous and the like.
  • the present disclosure is based on the unexpected discovery that reducing physiological bloodstream pH in a subject is useful in treating, ameliorating, and preventing many conditions and diseases and symptoms thereof in a subject in need.
  • the invention provides a stable therapeutic composition that can be administered to a subject in need thereof, in order to provide the requisite shift in blood pH.
  • FIG. 1 depicts a diagram of the chemiosmotic gradient potential of hydrogen ions in a normally functioning mitochondria in a mammalian cell.
  • blood and interstitial fluid typically has a pH of around 7.4
  • the intracellular fluid within a cell has a pH of around 7.28
  • intermembrane space of a mitochondria within the cell has a pH of around 6.88.
  • Ionic pumps concentrate H + ions in the intermembrane space of the mitochondria, resulting in a large H + gradient between the intermembrane space and mitochondrial matrix across the inner membrane.
  • the concentrations of other ionic species are also manipulated to create an electrochemical gradient across the various membranes, and intramitochondrial Ca 2+ in particular is important for managing the flow of H + ions within the mitochondria.
  • Hydrogen ions flow across the inner membrane into the mitochondrial matrix through ATP synthase, creating ATP from ADP.
  • the electron transport chain is used to pump the H + ions back across the inner membrane to maintain the proton gradient.
  • a small percentage of electron transfer occurs directly to oxygen, leading to free-radical formation, which contributes to oxidative stress and may result in membrane damage if insufficient antioxidants are present.
  • FIG. 2 depicts a diagram of the chemiosmotic gradient potential of hydrogen ions in a mitochondria in a mammalian cell with a dysfunctional metabolism, as may occur after a prolonged exposure to a poor diet, or lack of exercise.
  • the blood, interstitial space, and intracellular fluid have undergone acidotic shifts, i.e., increased the concentration of H + ions and reduced the pH.
  • the pH in the mitochondrial matrix is increased from normal due to membrane leaks or reduced H+ ion pumping action from the electron chain transport. As a result, the net H + electrochemical gradient available for the formation of ATP is reduced.
  • the cell and mitochondria must increasingly rely on other ionic species to provide the necessary electrochemical gradient on demand, such as through higher than normal concentrations of Ca 2+ within the intermembrane space “pushing” hydrogen ions across the inner membrane and a higher concentration of Cl ⁇ within the mitochondrial matrix “pulling” the hydrogen ions.
  • This dysfunctional ionic balance results in increased development of superoxidative species and increased membrane damage, and the metabolism of the cell slows down as a result. This reduces the amount of available ATP, causing a negatively reinforcing feedback loop that can lead to various adverse conditions and disorders.
  • a similar metabolic dysfunction occurs as a result of poor perfusion leading to a lactate burden, called metabolic acidosis in chronic state, which may be caused by, e.g., sepsis, multiple system atrophy (MSA), and ischemic conditions in peripheral limbs.
  • MSA multiple system atrophy
  • For individuals incurring a chronic lactate burden high blood levels of lactate steadily displace bicarbonate buffers to maintain acid-base homeostasis. A fraction of bicarbonate could then be removed by renal action to maintain homeostasis, and to reduce bloodstream bicarbonate levels.
  • chronic disturbances in electrolytes can shift the setpoint for bicarbonate retention to additionally reduce stores. Such forces would in turn make less bicarbonate accessible for intracellular retention and intracellular buffering, ultimately reducing intracellular H + stores. This reduction in H + stores would require more Ca 2+ to sustain a desired chemiosmotic gradient, leading to a dysfunctional ionic balance as described above.
  • Stable therapeutic compositions of the present disclosure reduce the physiological bloodstream pH in a subject, and maintain that reduction in physiological bloodstream pH for a duration of time, until renal and respiratory compensation processes negate the reduction, commonly followed by an alkaline “rebound”.
  • the compositions of the present disclosure are formulated such that the formulated pH is below the physiologic norm (i.e., below 7.4).
  • Bicarbonate concentration may, in some instances, be above physiologic norm (i.e., above 29 mM).
  • the sudden influx of H + ions, together with excess bicarbonate, and the manipulation of the electrochemical gradients that results allows for a return to normal mitochondrial metabolic processes, while other electrolyte, vitamin, and antioxidant support present in compositions of the present disclosure reduce the damage from oxidative stress.
  • compositions of the present disclosure include improvement of at least one of cardiovascular conditions, vasodilation, wound healing, vascular plaque, bicarbonate servicing, electrolyte economy, metabolic dysfunction, oxygen deficiency, Citric Acid Cycle, renal system operation, antioxidant dysfunction, angiogenesis, nitric oxide (NO) dysfunction, hormone function, and anemia.
  • cardiovascular conditions vasodilation, wound healing, vascular plaque, bicarbonate servicing, electrolyte economy, metabolic dysfunction, oxygen deficiency, Citric Acid Cycle, renal system operation, antioxidant dysfunction, angiogenesis, nitric oxide (NO) dysfunction, hormone function, and anemia.
  • the compositions of the present invention are suitable for improvement of cardiovascular conditions, by reducing or removing vascular plaques.
  • lipid dysfunction lipid dysfunction
  • nitric oxide dysfunction excessive ROS
  • smooth muscle contains several sources of ROS, which have been shown to function as important signaling molecules in the cardiovascular system.
  • the elevated ROS signals to the smooth muscles to accrue in the arteries, as though recruited to fill wounds that do not actually exist.
  • compositions of the invention are able to reduce or reverse vascular plaque by correcting or improving at least one of, nitric oxide dysfunction (thereby restoring NO signaling), lipid dysfunction, eNOS dysfunction, reduction in smooth muscle recruiting, reduction of endogenous and exogenous reactive oxygen species (ROS), elevated Ca 2+ , or restoration of fatty acid metabolism.
  • the smooth muscles upon the introduction of an alkaline environment, the smooth muscles, in the absence of the ROS signal, recognize the absence of a wound, and consequently, they down-regulate, and begin to directionally orient towards their vasodilation and vasoconstriction tasks.
  • foam cells are signaled to release their lipids.
  • the acid-shifting action of the drug liberates atomic components of the mineral deposits, while magnesium in the composition of the invention aids in the prevention of plaque re-deposition, to reduce the hardening of the arteries from the mineral deposit components.
  • the compositions of the present invention are suitable for preventing or minimizing hypoxia in a subject.
  • the lack of sufficient oxygen reaching cells or tissues in a subject can occur even when blood flow is normal. This can cause many serious, sometimes life-threatening complications.
  • Use of the compositions of the invention enable the resolution or improvement of conditions commonly associated with hypoxia, such as, for example, heart attack, cardiovascular problems, lung conditions, concussive cascade, reperfusion injury, myocardial infarction, hypoxia associated with diabetes, tissue trauma, and the like. Many of these conditions are associated with vasoconstriction.
  • the composition can counteract such vasoconstriction by promoting vasodilation via at least one of three pathways, namely endothelin, prostacyclin, or NO-soluble guanylyl cyclase (NO-sGC).
  • endothelin the compositions elevate Mg 2+ in the bloodstream to antagonize Ca 2+ . This blocks Ca 2+ from potentiating vasoconstriction, allowing the arteries to relax and dilate.
  • the compositions also provide metabolic corrections to reduce metabolic sources of ROS, and reduce the presentation of endothelin stimulants at the cell surface, thereby reversing Ca 2+ overstimulation.
  • niacinamide in the composition elevates adenosine 3′, 5′-cyclic monophosphate (cAMP) activity, which completes prostacyclin potentiation towards vasodilation.
  • cAMP adenosine 3′, 5′-cyclic monophosphate
  • the compositions of the invention provide a gradient of H + flowing into the cells to promote Ca 2+ efflux, which corrects elevated Ca 2+ presentation.
  • One effect of high levels of Ca 2+ is the elevation of caveolin. As the caveolin elevate, they take residence in the caveoli on the cell surface, causing the displacement of eNOS, which migrates to the Golgi system.
  • the combination of low ROS and low intracellular Ca 2+ achievable using the composition of the invention allows eNOS, to return from the Golgi to the cell membrane, thereby to restoring eNOS's ability to promote vasodilation.
  • the bloodstream pH shifts, promoting NO release via the NO-sGC pathway, and promoting vasodilation.
  • renal responses to rebalance pH produce a second “pH shift” towards alkaline, once again stimulating NO/NO-sGC vasodilation to extend the duration of the effect.
  • the compositions of the invention achieve both of these things, enabling the rapid resolution of the Ca 2+ overburden and the corresponding metabolic crisis.
  • the composition adjusts the pH of the bloodstream, acidifying it, and in doing so, causes H + to enter through the Na + /H + exchange route. As the H + enters, it pushes Na + out.
  • the composition of the invention promotes vessel vasodilation to improve blood flow. With this increased blood flow comes increased oxygen, entering, which enables the creation of ATP through aerobic metabolism.
  • the composition also elevates Mg 2+ in the bloodstream. The increased Mg 2+ facilitates the transport of the ATP, as Mg-ATP, to the Na + /K + ATPase, providing the stimulus to push Na + out.
  • the steady biasing towards alkaline and low ROS promotes positive rebalancing of electrolytes and pH in the cytosol, organelles, lysosomes, peroxisomes, calcium status, magnesium status and ROS status within the cell. Additionally, it changes the cellular economy to restore potassium and bicarbonate, while at the same time reducing intracellular calcium.
  • composition of the invention makes the composition useful for wound care. It was unexpectedly discovered that use of the compositions of the invention may provide wound recovery even in subjects who have exhausted conventional treatment methods, including those with gangrenous presentation, or chronic, diabetic or traumatic wounds. Metabolic changes are among the effects observed following trauma injury and surgical trauma. These include inflammatory responses, which trigger a constriction of blood flow to the affected regions. While this advantageously minimizes blood loss at the site of an open wound or internal bleed, it may impair healing by promoting a hypoxic intracellular environment. In trauma situations where bleeding risk is absent or reduced (for example by compression), it may be desired to suppress the inflammatory response, to avoid secondary injuries from hypoxia.
  • compositions of the invention are also capable of correcting key metabolic aberrancies that are present in wounds.
  • compositions may, for example improve at least one of restoring acid-buffer status and correction of elevated Ca 2+ ; reducing metabolic sourced ROS; correcting acidosis; correcting over-active iNOS and restoration of eNOS and nNOS function; promotion of beneficial angiogenesis after eNOS is corrected; and suppression of iNOS promoted aberrant angiogenesis, all of which are important for wound care.
  • H + also administrates acetylcholine uptake, which is part of muscle support, and is a part of the cerebellum control process, and ATP is relevant for all of these systems
  • disorders of the central nervous system are another treatment target.
  • action to resolve intracellular acid, calcium accrual, reduced ROS, and increased Mg are factors that can enhance function in the peroxisome, to better maintain catalase antioxidant supply, and additionally support the lipid modeling required for myelin maintenance of nerve sheaths.
  • the reduction in physiologic bloodstream pH caused by the composition of the invention may be minimal, or not observed, due to the particular formulation of an administered composition, the rate at which a composition is administered, or both.
  • the therapeutic benefits described herein may still be achieved due to the net elevation of bicarbonate concentration that occurs.
  • the body prioritizes retention of, and augmentation of, the buffer components (e.g., bicarbonate), as acid balancing processes proceed.
  • the buffer components e.g., bicarbonate
  • Such an “alkaline rebound” may result in bloodstream pH overshooting slightly for a net alkaline stabilization relative to the starting pH.
  • the “alkaline rebound” achieves a higher residual concentration of intercellular and bloodstream buffer components, including bicarbonate.
  • the system may regulate to a final pH equivalent to that present prior to treatment, but with bloodstream buffering, with regard to acidic species, being increased.
  • the bloodstream pH may settle to be more acidic than prior to the treatment, yet while a variety of aforementioned exchange phenomena are promoted.
  • co-administration of acid and buffer are key to limiting the H + efflux rate, while the intracellular calcium correction is achieved.
  • compositions of the present invention are suitable for increasing nitric oxide synthase (NOS) in a subject.
  • the pH biasing and increase in bicarbonate concentration as provided by compositions of the present disclosure may also restore endothelial and neuronal NOS, leading to a selective increase in nitric oxide production.
  • Nitric oxide is a gaseous signaling molecule with a role in, e.g., hemostasis, smooth muscle (particularly surrounding vasculature), neuronal signaling, and in the gastrointestinal tract.
  • NO has been implicated in a variety of physiological systems, and the increased levels resulting from administration of the compositions described herein may serve a role in providing the therapeutic benefits described herein.
  • NO may play a role in regulating intraocular pressure via the trabecular meshwork.
  • NO stops the aberrant perpetuation of smooth muscle recruitment, foam cell accrual and lipid storage, and collagen deposition, and it may ultimately lead to reversal of plaque damage and a return of the vascular section to physiological norms.
  • the compositions of the present invention are suitable for reducing lactate burden in a subject in need thereof.
  • lactate burden means any physiological condition characterized by elevated lactate levels. This may include, for example and without limitation, chronic lactate burdens such as acidosis, sepsis, and MSA, or acute lactate burdens such as may occur during and after physical exertion such as exercise. Lactate circulating oxygen debt burden that is retained in muscles, can be stimulated to be released by bicarbonate, and subsequently metabolized thus lowering the subject's lactate burden. The ability to eliminate lactate burden is important for a subject who has had, for example, an organ transplant.
  • the citrate must be metabolized. This metabolization can induce a lactate burden in those individuals. Additionally, lactate burden is a component of sepsis and a chronic burden in diabetics. In the above instances, as well as in others involving a lactate burden, the use of the compositions of the invention may reduce that burden.
  • the compositions of the present invention are suitable for reducing acidosis in a subject in need thereof, by administering to the subject the composition of the invention.
  • One of the metabolic effects of trauma is the suppression of insulin, resulting in a reduction of the normal anabolic effect of insulin towards an increase in catabolic effects. This leads to a shift towards free fatty acids as the primary source of energy, with triglycerides providing 50 to 80% of the energetic need. Reducing the catabolic response encourages faster healing after surgery. These same mechanisms are in play in the diabetic patient, and become a larger challenge as subjects progress in their metabolic dysfunction.
  • the composition of the invention facilitates Ca 2+ correction, and enhances B-vitamin servicing and ascorbic acid anti-oxidant servicing via elevated presentation. Additionally, acid burden is reduced, promoting an alkaline bias. Elevated HCO 3 ⁇ buffer levels also serve to preserve this alkaline bias.
  • the elements of metabolism referenced above also affect insulin management. For example, insulin release is stimulated from the pancreas when a signal of elevated Ca 2+ is released to the bloodstream. For Ca 2+ to be released to the pancreas, hydrogens must be created, through incomplete metabolism, to displace Ca 2+ from the cytosol to the bloodstream. As noted herein-above, the Na + /K + ATPase must be served with Mg 2+ and ATP to facilitate the flooding Na + to the bloodstream to ultimately stimulate the Na + /Ca 2+ exchanger to release Ca 2+ to the bloodstream. Additionally, for sensing of elevation to occur, the background level of Ca 2+ in the bloodstream needs to be low enough for the pancreas to observe the change.
  • ROS such as peroxide
  • Mg 2+ can promote insulin function, when presented at low levels, and prevent presentation and action of insulin when presented at high levels.
  • correction of acidosis and enhancement of Mg 2+ are key to restore insulin management. So too are suppression of ROS (e.g., H 2 O 2 ) through antioxidant support and facilitation of TCA and ECT function to achieve near-complete oxidation of Acetyl-CoA to CO 2 and H 2 O.
  • the composition of the invention is a stable therapeutic composition that has been formulated to make it suitable for intravenous administration to a subject.
  • the composition contains an intravenous buffer solution, containing at least one pharmaceutical grade acid, and at least one pharmaceutical grade pH buffering agent.
  • the acid and buffer solution are present in a sterile aqueous solution.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 3000 mmol/L when administered to a subject.
  • the acid and base are selected so that they are able together, to provide a buffer solution having a pH of between 4 and 7.7.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 80 mmol/L to 3000 mmol/L when administered to a subject, where the buffer solution is effective to provide a buffer solution pH of less than 5.5.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 100 mmol/L to 2000 mmol/L when administered to a subject, where the buffer solution is effective to provide a buffer solution pH of less than 5.5.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 200 mmol/L to 1000 mmol/L when administered to a subject, where the buffer solution is effective to provide a buffer solution pH of less than 5.5.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 40 mmol/L to 3000 mmol/L when administered to a subject, where the buffer solution is effective to provide a buffer solution pH of less than greater than or equal to 5.5.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 60 mmol/L to 2000 mmol/L when administered to a subject, where the buffer solution is effective to provide a buffer solution pH of less than greater than or equal to 5.5.
  • the concentration of the pharmaceutical grade acid and the pharmaceutical grade pH buffering agent in the buffer solution is sufficient to provide a total titratable acid content of from 80 mmol/L to 3000 mmol/L when administered to a subject, where the buffer solution is effective to provide a buffer solution pH of less than greater than or equal to 5.5.
  • compositions of the present disclosure comprise an acid that provides an amount of H + ions to decrease the physiological bloodstream pH in a subject. Without being bound to any theory, it is believed compositions of the present disclosure increase the H + gradient in various cellular environments, including, e.g., mitochondria.
  • This increased mitochondrial H + gradient drives higher production of ATP and, through other physiological homeostatic systems, causes changes in concentration gradients of the cellular membranes which in turn rebalances physiological ions such as sodium, magnesium, potassium, and calcium.
  • physiological ions such as sodium, magnesium, potassium, and calcium.
  • an increased H + gradient in the bloodstream may stimulate calcium pumps in cellular membranes, thereby increasing intracellular H + and reducing intracellular Ca 2+ .
  • the concentration gradients of sodium, magnesium, and potassium are also affected.
  • compositions of the present disclosure are sufficient to reduce the bloodstream pH of a subject by a small, moderate, or large amount.
  • the amount of acid in a composition of the present disclosure is sufficient to reduce the bloodstream pH of a subject by 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.15, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, or 1.1, or more.
  • the reduction in pH may also be expressed by the desired pH level of the bloodstream after administration of a composition of the present disclosure, e.g., 7.2.
  • a composition of the present disclosure comprises sufficient acid to reduce the bloodstream pH of a subject to 7.3, 7.2, 7.1, 7.0, 6.9, 6.8, 6.7, 6.6, 6.5, 6.4, or 6.3.
  • a reduction of bloodstream pH to below 6.3 is not typically advised, as it may pose a cell health risk and threaten the integrity of cellular phospholipid bilayers.
  • a “reduction” in pH provided by administration may still result in a bloodstream pH exceeding 7.4.
  • administration of a composition of the present disclosure may shift the physiological pH from 7.7 to 7.5.
  • compositions of the present disclosure may contain one or more pharmaceutical grade acids.
  • compositions of the present disclosure comprise a mixture of one or more pharmaceutical grade acids.
  • Acids may include any physiological acceptable acid, including, without limitation, hydrochloric acid, ascorbic acid, citric acid, lactic acid, phosphoric acid, or combinations thereof.
  • the pH of a composition of the present disclosure may be between about 4 and 7.7. In some embodiments, the pH of a composition of the present disclosure is between about 6.1. In embodiments where the pH of the composition is very low, the rate of administration may have to be managed to avoid tissue damage adjacent to the injection site as dilution is effected in the bloodstream.
  • compositions of the present disclosure comprise a pH buffering agent.
  • a pH buffering agent is a weak acid or base that is used to maintain the pH of a solution near a desired value.
  • Compositions of the present disclosure comprise a pH buffering agent such that the reduction in bloodstream pH may be sustained for a desired duration.
  • the pH buffering agent may comprise a conjugate acid or a conjugate base.
  • the pH buffering agent may comprise any physiological acceptable buffering agent, including, without limitation, sodium bicarbonate, a phosphate buffer, citrate buffer, or a synthetic buffer creating specific alkaline conditions (e.g., tris-hydroxymethyl amino methane), or combinations thereof.
  • the buffer capacity of a solution is a measure of the solution's ability to resist pH change, i.e., to maintain a specific pH level.
  • acid-base homeostasis relates to the proper balance of acids and bases in extracellular fluids, i.e., the pH of the extracellular fluid.
  • the pH of plasma is approximately 7.4 and is tightly maintained around that value by three interconnected systems: 1) buffering agents, including bicarbonate, phosphate, and proteins), 2) the respiratory system, which impacts the partial pressure of carbon dioxide in blood plasma, and 3) the renal system, which excretes waste acids and bases.
  • compositions of the present disclosure comprise a pH buffering agent in order to maintain the desired bloodstream pH level below the typical pH value of about 7.4 in the face of pressures exerted by the physiological systems that regulate acid-base homeostasis.
  • compositions of the present disclosure comprise a pH buffering agent in an amount sufficient to maintain the reduction in bloodstream pH, or to maintain the desired pH level, for a duration of 1 minute to 1 week.
  • the desired duration of the reduced bloodstream pH level will depend on the particular indication being treated as well as the individual being treated. In some embodiments, a small, moderate, or large buffer capacity may be desired.
  • a small quantity of drug and/or a slow administration of a drug product could stimulate compensatory processes that can be respiratory or renal, so as to mitigate observable acid shifting potential, but having stimulated respiratory and renal activity.
  • a blood stream response may be neutral or may tend toward alkaline.
  • administration of a high dose, and/or a dose with a fast administration rate, such as a bolus or fast IV drip could introduce the acid and overwhelm the compensatory processes to yield an observable downstream pH toward acidic.
  • a stimulus would commonly be expected to be followed by a rebound of bloodstream pH towards alkaline throughout the treatment or post-treatment.
  • the outcome resulting from a given dose level and/or administration rate may be different from patient to patient and from administration to administration as the patient's health, electrolytic status, pH status and compensatory process status evolve.
  • Different buffer capacities may be sufficient to maintain the reduction in bloodstream pH for a duration of 1 minute to 1 week.
  • the buffer capacity may also be expressed in molar equivalent of common buffers, such as bicarbonate.
  • the composition has a buffer capacity between 0.1 mM HCO 3 ⁇ equivalent and 1200 mM HCO 3 ⁇ equivalent. In other embodiments, the buffer capacity is between 0.1 mM HCO 3 ⁇ equivalent and 10 mM HCO 3 ⁇ equivalent. In some embodiments, the buffer capacity is between 10 mM HCO 3 ⁇ equivalent and 50 mM HCO 3 ⁇ equivalent. In some embodiments, the buffer capacity is between 10 mM HCO 3 ⁇ equivalent and 1000 mM HCO 3 ⁇ equivalent. In some embodiments, the buffer capacity is between 50 mM HCO 3 ⁇ equivalent and 800 mM HCO 3 ⁇ equivalent.
  • the buffer capacity is between 100 mM HCO 3 ⁇ equivalent and 600 mM HCO 3 ⁇ equivalent. In some embodiments, the buffer capacity is between 200 mM HCO 3 ⁇ equivalent and 550 mM HCO 3 ⁇ equivalent. In some embodiments, the buffer capacity is between 20 mM HCO 3 ⁇ equivalent and 100 mM HCO 3 ⁇ equivalent. In other embodiments, buffer capacity may be expressed by the molar concentration of HCO 3 ⁇ , or other common buffers. For example, in some embodiments, the molar concentration of HCO 3 ⁇ may be between 0.01 molar and 10 M. In other embodiments, the molar concentration of HCO 3 ⁇ may be between 0.5 and 2 M.
  • the present disclosure provides a composition having a pH below physiological pH (i.e., below 7.4) and an HCO 3 ⁇ concentration above physiological levels (i.e., above 29 mM).
  • the pH of the composition may be between 4 and 7.7 and the HCO 3 ⁇ concentration may be between 30 mM and 2 M).
  • the pH of the composition may be between 5.5 and 7.4.
  • the pH of the composition may be around 6.
  • FIG. 5 shows a diagram of the amplitude and duration of an acid state shift caused by different formulations of compositions of the present disclosure.
  • the gray lines, both solid and dotted depict a smaller acid shift, i.e., a composition with a lower concentration of H + ions. Again, the buffer capacity between these compositions varies such that the acid shift caused by the composition depicted by the dotted gray line is maintained for a shorter duration.
  • Compositions of the present disclosure may be designed along these two spectrums, amplitude of shift and duration of shift, according to desired therapeutic properties and administration schedules.
  • the present disclosure provides a stable therapeutic composition
  • a buffer solution comprising a pharmaceutical grade base and at least one pharmaceutical grade conjugate acid, wherein the buffer solution is sufficient to raise the physiological bloodstream pH of a subject by 0.1 to 1.1, and wherein the buffer solution has a buffer capacity sufficient to sustain the elevation of the physiological bloodstream pH.
  • the buffer capacity may be sustained for a period of time for example 1 minute or 1 week.
  • the compositions may further comprise vitamins, salts, acids, amino acids or salts thereof, and stabilized oxidative species.
  • compositions of the present disclosure may further comprise salts to provide sources of physiological relevant ionic species, such as Na + , K + , Mg 2+ , Cl ⁇ , PO 4 3 ⁇ , or Ca 2+ .
  • physiological relevant ionic species such as Na + , K + , Mg 2+ , Cl ⁇ , PO 4 3 ⁇ , or Ca 2+ .
  • These may include, without limitation, sodium chloride, disodium phosphate, potassium chloride, monopotassium phosphate, magnesium chloride, and calcium chloride.
  • the compositions may further comprise other trace elements and their salts, including, but not limited to, selenium, copper, chromium, iodine, fluoride, zinc, manganese, molybdenum, and iron.
  • Sodium ions are required in relatively large concentrations for normal physiological functioning. It is the major cation of the extracellular fluid. It plays an important role in many physiological processes, including the regulation of blood volume, blood pressure, osmotic equilibrium, and pH, as well as the generation of nerve impulses.
  • Potassium ions are the major cation of intracellular fluid, and, with the sodium ions of the extracellular fluid, is a primary generator of the electrical potential across cellular membranes. Accordingly, it plays a significant role in normal functioning, and is critical in such body functions as neurotransmission, muscle contraction, and heart function.
  • Ca 2+ ions are likewise important to many physiological processes.
  • Ca 2+ ions are one of the most widespread second messengers used in signal transduction.
  • Ca 2+ ions may regulate several signaling pathways which cause smooth muscles surrounding blood vessels to relax.
  • Dysfunction within Ca 2 -activated pathways can lead to an increase in tone caused by unregulated smooth muscle contraction. This type of dysfunction can be seen in cardiovascular diseases, hypertension, and diabetes.
  • Magnesium ions are required in relatively large concentrations in normal metabolism. It is recognized that deficiency of magnesium is rare unless it is accompanied by severe losses in other electrolytes such as in vomiting and diarrhea. It is however frequently recognized as deficient in the modern diet with symptoms such as muscle tremors and weakness. This mineral is important in many enzymatic reactions and will stabilize excitable membranes. Administered intravenously, magnesium may produce an anesthetic action and this is indirect evidence of its action on the vascular wall endothelial component to stabilize and normalize the surface of the vascular wall.
  • a composition of the present disclosure comprises Na + at a concentration between 0.1 mM and 1 M. In other embodiments, a composition of the present disclosure comprises K + at a concentration between 0.0 mM and 1 M. In some embodiments, a composition of the present disclosure comprises Mg 2+ at a concentration between 0.1 mM and 1 M. In other embodiments, a composition of the present disclosure comprises Ca 2+ at a concentration between 0.1 mM and 1 M.
  • compositions of the present disclosure may include these species to aid in the restoration of normal physiological conditions and concentrations.
  • high intracellular Ca 2+ may be restored to a lower level as offset by Mg 2+ , K + , and H + , which may lead to NOS presentation in the cytosol and restoration of NO levels.
  • the compositions described herein may include vitamins and vitamers, which is a substance(s) that has vitamin-like activity. Vitamins selected from the group consisting of the water soluble and lipid soluble group, and a combination of two or more thereof may also be added to the pharmaceutical composition.
  • the pharmaceutical composition includes ascorbic acid. Ascorbic acid is included as a strong antioxidant component and to maintain the structural integrity of connective tissue, including epithelial basement membranes and to promote wound healing. It may also play a distinct role as an agent with strong anti-inflammatory actions. The oxidized form of the vitamin, dehydroascorbic acid, has been shown to transfer intracellularly where some of it is reduced within the cell via action of glutathione.
  • a composition of the present disclosure comprises dehydroascorbic acid, an oxidized form of ascorbic acid that is actively imported into the endoplasmic reticulum of cells via glucose transporters. Presentation of dehydroascorbic acid can also stimulate production of glutathione in the liver, which facilitates recycling of dehydroascorbic acid into ascorbic acid. Thus, dehydroascorbic acid indirectly enhances intracellular antioxidant resources.
  • Dehydroascorbic acid may be present via direct inclusion of pharmaceutical grade dehydroascorbic acid, or by conversion of ascorbic acid via contact with a reactive oxygen species such as HOCl, H 2 O 2 , or OCl.
  • the B Group of Vitamins has been shown to be important in human food intake, and plays an important role acting as co-enzymes in cellular metabolism and energy production.
  • the entire B group of vitamins may be included in the formulation to address any deficiencies in the patient population to be treated.
  • the B group vitamins are found to occur naturally together in foods and are generally included comprehensively for this reason.
  • the B group includes: 1) Thiamine (B1), which plays an important role in energy production within the cell, specifically as co-enzyme in metabolism of carbohydrates. At least 24 enzymes are known to use thiamine as a co-enzyme; 2) Riboflavin (B2) in the form of flavin mononucleotide and flavin adenine dinucleotide are part of all dehydrogenase enzymes.
  • Niacinamide (B3) is included, as part of the B group of vitamins as deficiency syndromes in clinical pellagra are well known clinical manifestations of deficiencies.
  • the deficiency states of this vitamin are associated with intestinal diseases and alcohol misuse. It also occurs in diabetes mellitus and carcinoid syndrome.
  • the active forms of this vitamin include the nicotinamide dinucleotides NAD and NADP, which are the co-enzymes and co-substrates for numerous dehydrogenases responsible for oxidation-reduction systems within the human cell, which are indispensable for energy production.
  • nicotinic acid from the administered nicotinamide in the formulation produce nicotinic acid possessing additional actions not shared by nicotinamide, such as inhibition of cholesterol synthesis; 4) Calcium D-Pantothenate (B5), pantothenic acid forms a major part of the molecule of co-enzyme A, which is important in the energy producing metabolic cycles in the mitochondria of all cells.
  • Kynureminase which is an enzyme used to identify pyridoxine deficiencies, loses its activity when pyridoxine is not present and may result in secondary nicotinic acid deficiency as a result of lack of the kynureminase conversion of nicotinic acid from tryptophan.
  • Cyanocobalamin (B12) is used because of the frequent reports of mal-absorption of cyanocobalamin, caused by poor dietary habits, senescence, and certain drugs (metformin) used as a hypoglycemic agent in diabetes mellitus.
  • This vitamin is essential for normal erythropoiesis to occur, and recent findings have also implicated this vitamin with improvement of neuronal transmission in motor neuron disease. (Rosenfeld, Jeffrey and Ellis, Amy, 2008, Nutrition and Dietary Supplements in Motor Neuron Disease, Phys Med Rehabil Clin N Am., 19(3):573-589).
  • Vitamin K is a fat-soluble vitamin. There are two naturally occurring forms of the vitamin. Vitamin K1 is the dietary Vitamin K and is abundant in green leafy vegetables, whereas vitamin K2 is present in tissues. Vitamin K2 is synthesized by bacteria. It is found mainly in fermented products like fermented soybeans, cheese, curds and to some extent also in meat and meat products (Thijssen, H. H., M. J. Drittij-Reijnders, and M. A. Fischer, 1996, Phylloquinone and menaquinone-4 distribution in rats: synthesis rather than uptake determines menaquinone-4 organ concentrations, J Nutr 126:537-43). Vitamin K2 is found in animals as menaquinone.
  • vitamin K It is the human activated form of vitamin K and is said to promote the healing of bone fractures. It is essential for the carboxylation of glutamate residues in many calcium binding proteins such as calbindin and osteocalcin. These proteins are involved in calcium uptake and bone mineralization.
  • vitamin K1 for, but not for vitamin K2.
  • a typical therapeutic oral dose for vitamin K2 for osteoporosis is 45 mg/day.
  • a much higher level of vitamin K is needed for complete gamma-carboxylation of osteocalcin (Booth, S. L., and J. W. Suttie, 1998, Dietary intake and adequacy of vitamin K, J. Nutr 128:785-8).
  • Vitamin K deficiency is associated with reduced hip bone mineral density and increased fracture risk in healthy elderly women. Animal studies have shown that the most potent form of vitamin K is vitamin K2, which was administered to rats at 0.1 mg/kg orally (Akiyama, Y., K. Hara, A. Matsumoto, S.
  • Vitamin K2 in the form of menaquinone-4, is the most biologically active form. It has been extensively studied in the treatment of osteoporosis. In one of these studies, 241 osteoporotic women were given 45 mg/day vitamin K2 and 150 mg elemental calcium. After two years, vitamin K2 was shown to maintain lumbar bone mineral density, significant lower fracture incidence (10% versus 30% in the control group (Shiraki, M., Y. Shiraki, C. Aoki, and M. Miura, 2000, Vitamin K2 (menatetrenone) effectively prevents fractures and sustains lumbar bone mineral density in osteoporosis, J Bone Miner Res 15:515-21).
  • Vitamin K2 may inhibit the calcification of arterial plaque.
  • animal studies involving rats found high dose of vitamin K2 (100 mg/kg body weight daily) inhibited the increase in calcium in both kidneys and aorta induced by megadose of synthetic vitamin D (Seyama, Y., M. Horiuch, M. Hayashi, and Y. Kanke, 1996, Effect of vitamin K2 on experimental calcinosis induced by vitamin D2 in rat soft tissue, Int J Vitam Nutr Res 66:36-8).
  • a similar study was conducted with rabbits.
  • Vitamin K2 (1-10 mg/kg daily for 10 weeks) inhibited the atherosclerotic plaque progression in the aorta and pulmonary arteries (Kawashima, H., Y. Nakajima, Y. Matubara, J. Nakanowatari, T. Fukuta, S. Mizuno, S. Takahashi, T. Tajima, and T. Nakamura, 1997, Effects of vitamin K2 (menatetrenone) on atherosclerosis and blood coagulation in hypercholesterolemic rabbits, Jpn J Pharmacol 75:135-43).
  • Vitamin K2 was also seen to reduce total cholesterol levels, lipid peroxidation, ester cholesterol deposition in the aorta and factor X activity in plasma compared to the control group.
  • a composition of the present disclosure comprises one or more of the vitamins or vitamers above.
  • a composition may comprise one or more of the vitamins or vitamers above in amounts between 1 ⁇ g and 1,000 mg per dose.
  • a composition of the present disclosure may further comprise antioxidant compounds.
  • antioxidant compounds may include, but are not limited to, nonenzymatic compounds such as tocopherol (aTCP), coenzyme Q10 (Q), cytochrome c (C) and glutathione (GSH), and enzymatic components such as manganese superoxide dismutase (MnSOD), catalase (Cat), glutathione peroxidase (GPX), phospholipid hydroperoxide glutathione peroxidase (PGPX), glutathione reductase (GR); peroxiredoxins (PRX3/5), glutaredoxin (GRX2), thioredoxin (TRX2) and thioredoxin reductase (TRXR2).
  • a composition may comprise one or more of the antioxidant compounds above in amounts between 1 ⁇ g and 1000 mg per dose.
  • a composition of the present disclosure may further comprise a stabilized oxidative species.
  • the stabilized oxidative species may be, without limitation, one or more of H 2 O, O 2 , H 2 O 2 , Cl 2 O and H 3 O.
  • adjuncts may include selenium and/or selenocysteine at concentrations of 60 to 90 ⁇ g per dose.
  • Other adjuncts may also include other trace elements and their salts, including, but not limited to, copper, chromium, iodine, fluoride, zinc, manganese, molybdenum, and iron.
  • compositions of the present disclosure may be formulated by combining pharmaceutical grade compounds into a stable therapeutic composition.
  • Compounds may be added in desired amounts to a vessel, with water added to complete a final volume.
  • a composition of the present disclosure comprises a final volume of between 5 mL and 500 mL.
  • a composition comprises a final volume of about 250 mL.
  • the composition may be provided in 20 mL vials.
  • a composition of the present invention may be further diluted prior to administration. For example, a 20 mL vial may be diluted with saline to a 100 mL dispensed volume for administration.
  • the liquid formulation may be reduced to dry solid via lyophilization. The lyophilized formulation may then be reconstituted to a particular volume prior to administration.
  • Table 1 shows various formulations of the composition according to exemplary embodiments of the present disclosure per 20 mL vial:
  • compositions in Table 1 may be varied from the listed values by plus or minus 1%, 2%, 5%, or 10% according to therapeutic need.
  • the compositions of Table 1 may also further comprise additional components as described above according to therapeutic need.
  • compositions of the present disclosure may be stabilized to enhance shelf life.
  • the compositions may be stabilized by suitable techniques as known to those of ordinary skill in the art, including, but not limited to, freezing, lyophilization, use of UV or spectral blocking vials (e.g., amber vials), overfilling with stabilizing gases such as nitrogen, bubbling a stabilizing gas through the solution, separating reactive species into multiple vials to be combined upon use, and cold chain storage.
  • the acid and buffer components of a composition may be separated into two vials.
  • Other components of compositions of the present disclosure e.g., cyanocobalamin, calcium d-pantothenate, and/or others
  • the present disclosure provides methods of treatment.
  • the methods of the invention involve administering the composition of the invention to subjects in need thereof.
  • One embodiment of the invention is a method of treating or ameliorating a mitochondrial disorder, metabolic disorder, a condition associated with diabetes, a cardiovascular dysfunction, or an ocular condition in a subject in need thereof, by administering to the subject a stable therapeutic composition of the present disclosure.
  • Mitochondrial dysfunction characterized by a loss of efficiency in the electron transport chain and reductions in the synthesis of high-energy molecules, such as ATP, is a characteristic of aging, and essentially, of all chronic diseases.
  • mitochondrial dysfunction refers to a condition or disorder characterized by mitochondrial dysfunction, and includes, for example, neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Friedreich's ataxia, cardiovascular diseases, such as atherosclerosis and other heart and vascular conditions, diabetes and metabolic syndrome, autoimmune diseases, such as multiple sclerosis, systemic lupus erythematosus, and type 1 diabetes, neurobehavioral and psychiatric diseases, such as autism spectrum disorders, schizophrenia, and bipolar and mood disorders, gastrointestinal disorders, fatiguing illnesses, such as chronic fatigue syndrome and Gulf War illnesses, musculoskeletal diseases, such as fibromyalgia and skeletal muscle hyper
  • a “metabolic disorder” refers to diabetes, insulin resistance, glucose intolerance, hyperglycemia, hyperinsulinemia, obesity, hyperlipidemia, or hyperlipoproteinemia.
  • diabetes insulin resistance, glucose intolerance, hyperglycemia, hyperinsulinemia, obesity, hyperlipidemia, or hyperlipoproteinemia.
  • diabetes mellitus are intended to encompass both insulin dependent and non-insulin dependent (Type 1 and Type 2, respectively) diabetes mellitus, gestational diabetes, as well as pre-diabetes, unless one condition or the other is specifically indicated.
  • a “condition associated with diabetes” includes obesity, hypertension, hyperlipidemia, fatty liver disease, nephropathy, neuropathy, renal failure, retinopathy, diabetic ulcer, cataracts, insulin resistance syndromes and cachexia.
  • cardiovascular dysfunction includes conditions and diseases such as coronary heart disease, cerebrovascular disease, hypertension, peripheral artery disease, occlusive arterial disease, angina, rheumatic heart disease, congenital heart disease, heart failure, cardiac insufficiency, palpitations, supraventricular tachycardia, fibrillation, faintness, dizziness, fatigue, migraine, high levels of total blood cholesterol and/or LDL cholesterol, low level of HDL cholesterol, high level of lipoprotein, infections of the heart such as carditis and endocarditis, diabetic ulcer, thrombophlebitis, Raynaud's disease, anorexia nervosa, claudication and gangrene, atherosclerosis and peripheral artery disease.
  • Diseases and conditions that are especially suited for treating or ameliorating with a pharmaceutical grade buffer composition as described herein are peripheral artery disease and atherosclerosis.
  • the term “ocular condition” refers to pathological conditions pertaining to the eye, and may include, but is not limited to, glaucoma, macular degeneration, light sensitivity issues, calcific and collagen-based floaters, lens rigidity correction.
  • Another embodiment of the invention is a method of treating or ameliorating a dermatological condition by administering to the subject a stable therapeutic composition of the present disclosure
  • skin-related disorders, conditions and disease such as skin aging, wrinkles (including, e.g., laugh lines and wrinkles surrounding the eye), acne, photodamage, rosacea, scars, eczema, alopecia, hypertrophic scars, keloids, stretch marks or Striae distensae, psoriasis, pruritus, ehlers-danlos syndrome, scleroderma, post inflammatory hyperpigmentation, melasma, alopecia, poikiloderma of civatte, vitiligo, skin dyschromia, burns and blotchy pigmentation.
  • the present disclosure provides a method of modifying the metabolism of a subject, the method comprising administering to the subject a stable therapeutic composition comprising a buffer solution comprising at least one pharmaceutical grade acid and at least one pharmaceutical grade pH buffering agent, wherein the buffer solution is sufficient to reduce the physiological bloodstream pH of a subject by 0.01 to 1.1, and wherein the buffer solution has a buffer capacity sufficient to sustain the reduction of the physiological bloodstream pH of the subject for between 1 minute and 1 week.
  • a different embodiment of the invention provides a method of reducing lactate burden in a subject in need thereof, by administering to the subject the composition of the invention.
  • the reduction of lactate burden has been described extensively herein-above.
  • the present disclosure provides a method of reducing acidosis in a subject in need thereof, by administering to the subject the composition of the invention.
  • the reduction of acidosis has been described extensively herein-above.
  • the present disclosure provides a method of treating a central nervous system disorder in a subject in need thereof, by administering the composition of the invention.
  • central nervous system disorder means any neurological disorder affecting the structure or function of the brain or spinal cord.
  • the present disclosure provides a method of treating chronic wounds of a subject, by administering the composition of the invention. In some embodiments, the present disclosure provides a method for inducing accelerated wound healing in a subject, the method by administering a stable therapeutic composition of the present disclosure.
  • the present disclosure provides a method of enhancing mental or physical performance of a subject, by administering the composition of the invention.
  • compositions of the present disclosure include, but are not limited to, intravenous, intramuscular, or parenteral administration, oral administration, otic administration, topical administration, inhalation or otherwise nebulized administration, transmucosal administration and transdermal administration.
  • Compositions of the present disclosure may also be formulated for intravenous, bolus, dermal, oral, otic, suppository, buccal, ocular, or inhalation delivery.
  • the composition may also contain suitable pharmaceutical diluents and carriers, such as water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative, or synthetic origin. It may also contain preservatives, and buffers as are known in the art.
  • suitable pharmaceutical diluents and carriers such as water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative, or synthetic origin. It may also contain preservatives, and buffers as are known in the art.
  • the solution can also contain components to adjust pH, tonicity, stability, and the like, all of which is within the skill in the art.
  • the composition may be formulated in, e.g., liquid, gel, paste, or cream. In some embodiments, the composition may be administered via a topical patch.
  • the composition may be formulated in, e.g., liquid eye drops, or as a gel, paste, or cream to be applied to the surface of the eye and/or surrounding tissue.
  • the composition may be formulated in, e.g., ear drops.
  • a composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to peptide an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection Citrate Buffer pH 5.5, or other carriers, diluents and additives as known in the art.
  • an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection Citrate Buffer pH 5.5, or other carriers, diluents and additives as known in the art.
  • compositions of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additive known to those of skill in the art.
  • the pharmaceutical compositions are formulated for intravenous or parenteral administration.
  • compositions for intravenous or parenteral administration comprise a suitable sterile solvent, which may be an isotonic aqueous buffer or pharmaceutically acceptable organic solvent.
  • compositions for intravenous or parenteral administration can optionally include a local anesthetic to lessen pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form in a hermetically sealed container such as an ampoule or sachet.
  • the pharmaceutical compositions for administration by injection or infusion can be dispensed, for example, with an infusion bottle containing, for example, sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection, saline, or other solvent such as a pharmaceutically acceptable organic solvent can be provided so that the ingredients can be mixed prior to administration.
  • the duration of intravenous therapy using the pharmaceutical composition of the present invention will vary, depending on the condition being treated or ameliorated and the condition and potential idiosyncratic response of each individual mammal.
  • the duration of each infusion is from ⁇ 1 minute (e.g., bolus injection) to about 1 hour (intravenous delivery).
  • the infusion can be repeated within 24 hours.
  • a mammal can receive about 1 to about 25 infusions per day.
  • the number of infusions per day is 1 or 2.
  • the period between each infusion can be 1, 2, 5, 10, 20, 30, 40, 50 minutes, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 hours or more.
  • the administration may also be administered at any of a variety of cadences, including hourly, daily, weekly, monthly, quarterly, bi-annually, annually, etc., or any other particular timeframe depending on the condition to be treated and/or the response of each individual mammal.
  • a pharmaceutical composition of the present invention may be administered as a single event, or may be administered over week-long, multi-week, month-long, year-long, or multi-year durations, or for any other desired duration as may be warranted.
  • the infusions can be given one after another without a substantial period in between. In one embodiment, the infusion lasts about 45 minutes.
  • the dose may be repeated 2-3 times a week depending on the severity of the relative or absolute deficits of nutrients in the patient.
  • a clinical assessment may be necessary in order to establish the status, but can be limited to a review of medical history, subjective review of symptoms, the subjective opinion of the mammal when human or upon review of any specific deficits.
  • administration is alternated between two solutions: one acid shifting (AS) and one base shifting (BS) as described above.
  • AS acid shifting
  • BS base shifting
  • Alternating administration of AS/BS/AS/BS in various cadences would be expected to induce more pH swings from acidic towards basic or from basic towards acidic.
  • an exemplary administration profile may be a 5 minute AS administration followed by a 10 minute BS administration, repeated two times (i.e., 5/10/5/10).
  • Other exemplary administration profiles may be, e.g., 10/10/10/10 or 0.5/0.5/0.5/0.5.
  • Systemic formulations include those designed for administration by injection, e.g., subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection.
  • Useful injectable preparations include sterile suspensions, solutions or emulsions of the active compound(s) in aqueous or oily vehicles.
  • the compositions also can contain solubilizing agents, formulating agents, such as suspending, stabilizing and/or dispersing agent.
  • the formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, and can contain added preservatives.
  • the compound can be administered to a patient at risk of developing one of the previously described conditions or diseases.
  • prophylactic administration can be applied to avoid the onset of symptoms in a patient suffering from or formally diagnosed with the underlying condition.
  • the amount of compound administered will depend upon a variety of factors, including, for example, the particular indication being treated, the mode of administration, whether the desired benefit is prophylactic or therapeutic, the severity of the indication being treated and the age and weight of the patient, the bioavailability of the particular active compound, and the like. Determination of an effective dosage is well within the capabilities of those skilled in the art coupled with the general and specific examples disclosed herein.
  • Formulations can comprise other ingredients for the treatment of the organism as a whole.
  • an anti-oxidant additive and/or pro-oxidant additive can be present.
  • the latter may be an agent that acts as a preventive, while the former may be an agent that acts to treat a specific medical condition.
  • Efficacy of treatment may be determined by measuring biomarkers before, during, and/or after administration of a composition of the present disclosure, or before, during and/or after administration of a course of treatment using compositions of the present disclosure.
  • Exemplary biomarkers, and the indications for which they may be used, are shown in Table 2, and may include, e.g., A1Micro, tubular disorders and electrolyte imbalance; A2Macro, cerebral small vessel disease, liver fibrosis; ACE, high blood pressure, heart failure, diabetic nephropathy; Adiponectin, vascular disease, metabolic syndromes; Apo A-I, high density lipid particles; Apo A-II, HDL metabolism; Apo C-II, ischemic stroke, heart disease; Apo C-III, metabolic syndrome and hypertriglyceridemia; Apo H, type 2 diabetes, metabolic syndrome; AT-III, venous thrombosis, abnormal coagulation; B2M, peripheral arterial disease; BDNF, psychia
  • kits for administering the stable therapeutic composition of the present disclosure may contain the composition in a single vial or in more than one vial.
  • the vial can preferably be an injection vial with a membrane that is suitable for inserting a syringe to pull the solution from the vial or a soft I.V. infusion bag.
  • the composition of the invention is contained in the vial in a sterile aqueous solution.
  • the solution can be provided as a concentrated solution to which a diluent is added prior to administration.
  • the diluent can be sterile water.
  • the kit may further comprise a pre-filled container which contains the diluent.
  • a soft infusion bag is pre-filled with diluent.
  • the composition vial can contain a solution that is at a concentration which is suitable for injection without any dilution.
  • the solution for injection is isotonic. That is, the solution can contain salt, carbohydrates, such as glucose, NaHCO 3 or amino acids, such as glycine, and is isotonic with blood plasma.
  • the solution may be hypotonic so as to promote more rapid intracellular uptake or hypertonic so as to promote slower intracellular uptake.
  • the kit contains two vials.
  • the first vial at least one pharmaceutical grade acid in a sterile aqueous solution.
  • the first vial may contain pharmaceutical grade ascorbic acid, thiamine HCl, magnesium sulfate, cyanocobalamin, niacinamide, pyroxidine HCl, riboflavin 5′ phosphate, calcium D-pantothenate, and an aqueous solvent containing sodium chloride and water (for injection).
  • the second vial contains at least one pharmaceutical grade pH buffering agent in a sterile aqueous solution.
  • the second vial may contain pharmaceutical grade sodium bicarbonate and an aqueous solvent containing sodium chloride and water (for injection). The contents of the vials may be stored under refrigeration or under freezing conditions.
  • the kit may contain a container of a lyophilized powder that may be reconstituted prior to administration.
  • the lyophilized powder may be an isotonic solution.
  • kits described herein may further comprise instructions for use.
  • the instructions will, of course, depend upon the kit itself and whether a diluent is to be used or other components to be admixed with the pharmaceutical grade buffer solution prior to administration.
  • Blood has acid-base properties that can be notionalized as a solution having a physiological pH and buffer capacity.
  • therapeutic compositions designed to shift blood pH upon administration can be notionalized as solutions having a target pH and buffer capacity.
  • pH shifting of the bloodstream towards acidic conditions can be achieved via intravenous or intraarterial administration of an acid solution.
  • Alternative formulations having higher concentrations of buffer components have increased capacity to impede the restoration of bloodstream pH back towards physiologic.
  • Faster dissolution of calcified mineral forms can be achieved when the conditions are shifted from equilibrium at a given pH to lower pH levels.
  • C1-C4 examples drug products
  • HCl and NaHCO 3 examples acid and buffer components
  • Blood and Drug Compositions per Table 3 Below By design, these would have pH below physiologic and be comprised of buffer products as well.
  • C1 was designed to provide a small pH shift for a short time
  • C2 was designed to provide a small pH shift for a long time
  • C3 was designed to provide a large pH shift for a short time
  • C4 was designed to provide a large pH shift for a long time.
  • C1, C2, C3, and C4 were formulated, as was the blood surrogate, and the pH was measured. The pH was also calculated per the Henderson-Hasselbalch equation.
  • the compositions C1, C2, C3, and C4 were added to the surrogate blood. Again, a pH value was calculated, and a pH value was measured.
  • each of the example therapeutic solutions shift the bloodstream pH from physiologic norm conditions (e.g., 7.41 pH) to reduced pH (e.g., 7.31-6.70 pH). This is demonstrated through addition of the C1 (or 2, 3, 4) to the blood surrogate solution, as summarized in Table 3.
  • the therapeutic formulations with the lowest pH and/or larger buffer fraction are capable of imparting a larger shift in bloodstream pH.
  • calcium salts were submerged in therapeutically pH-shifted blood surrogate solution and dry weighed after select time intervals of submersion.
  • the blood surrogate was first exposed to a large quantity of calcium salt for an extended period while at pH 7.41 to establish equilibrium of the salt at the start pH. Then residual solid calcium salts were removed, leaving a blood surrogate solution that was near-saturated with the calcium salt at the 7.41 pH. Then C1 (or 2, 3, 4) formulations were added to the calcium saturated blood surrogate to reduce the pH. Then the 2 g pellets of the calcium salts were submerged in therapeutically pH-shifted solution and dry weighed at select time intervals to establish a rate of weight loss (Table 4).
  • compositions of the present disclosure depending upon and as dictated by the therapeutic needs of the patient.
  • features illustrated or described as being part of one embodiment may be used on another embodiment to yield a still further embodiment.
  • a Vial products were refrigerated at 40° F. prior to use, while B Vial products were stored at 70° F.
  • 100 ml of A Vial product was combined into a saline IV bag, and then 100 ml of B Vial product was combined into the IV bag.
  • the IV bag was hung from an elevation point, 18′′ above infusion point.
  • a catheter was inserted into the jugular vein of the subject.
  • Post-treatment sample were subjected to blood gas analysis (acid/base status, Oximetry, Electrolytes and Metabolites) * Note: Subject 1's “pre-treatment” venous blood samples for IDEXX analysis (hematology, chemistry, endocrinology and serology) were mistakenly sampled 60 minutes after treatment began. The results likely reflect post-dose changes in plasma volume, as large changes were observed for concentration-based markers (e.g., RBC, hematocrit).
  • concentration-based markers e.g., RBC, hematocrit
  • venous HCO 3 ⁇ was first measured to have a high value of 33.2 mmol/L, consistent with Cushing's disease. Over treatment, the values reduced to 31 mmol/L at the other time points, consistent with a flow to the intracellular or renal extraction. As shown in FIG.
  • venous sO 2 and pO 2 were observed to rise during this time, from low start levels at 55% sO 2 and 30 mm Hg pO 2 , consistent with an enhanced servicing of oxygen to tissues.
  • pCO 2 was seen to reduce, consistent with a reduction in metabolism, plasma volume expansion, or reduced hemoglobin affinity for CO 2 , with heightened affinity for O 2 .
  • venous sO 2 and pO 2 were again observed to rise during drug administration, along with a reduction in pCO 2 .
  • dose 5 provoked a response unlike doses 1 and 4, where venous pH was observed to drop towards acidic throughout the observation frame. This could be attributed to the more alkaline starting bias for the bloodstream.
  • venous sO 2 and pO 2 were observed to have higher start levels at 73% sO 2 and 37 mmHg pO 2 , constant with a more durable restoration of enhanced servicing of oxygen to tissues.
  • sO 2 and pO 2 were again observed to rise further during drug administration.
  • pCO 2 remained largely unchanged.
  • the difference in behavior at dose 5, relative to doses 1 and 4 is consistent with achievement of an enhanced homeostasis regarding acid/base status.
  • dose 5 provoked a response similar to dose 5 in Subject 2, where venous pH was observed to drop towards acidic throughout the observation frame.
  • venous sO 2 and pO 2 were observed to have relatively high start levels at 70% sO 2 and 34 mm Hg pO 2 , consistent with a biasing towards enhanced servicing of oxygen to tissues, relative to pre-treatment levels.
  • sO 2 and pO 2 responded to AS product infusion by dropping during drug administration, which is a stimulus that is recognized to have the potential to stimulate EPO release from the liver to promote RBC store supplementation. This difference in response could have been caused by the formulation differences between the AS and ASVM configurations. pCO 2 rose correspondingly during this time.
  • Subject 1 Dose 1, 4, 5 ASVM Data (Exhibited for completeness, Similar to Subject 2 and Subject 3):
  • Hb reduced from its start value, sometimes showing evidence of rebound during the observation period. This cannot be interpreted as hemolysis, as a rebound on this timescale would be impossible.
  • the change in Hb concentration is consistent with a change in blood volume, likely due to a change in plasma volume. Such exchange is required during exercise-like stimulus to maintain vascular pressure, under conditions of vasodilation, where vascular volume increases.
  • Lactate was observed to have a low presentation value, which further reduced in Dose 4, and elevated slightly in Dose 5. This is consistent with the lactate burden steadily dropping with successive doses, as improved perfusion increased aerobic metabolism, so as to resolve lactate debt. Elevated lactate was seen in Dose 5, despite s suspected plasma volume dilution. This is consistent with presentation of HCO3 ⁇ into muscles to release stored lactate.
  • Lyme Disease Equine - Multiplex The Lyme multiplex assay determines antibodies to three antigens, called outer surface proteins (Osp), of B. burgdorferi which have been shown to correlate with vaccinal antibodies, or acute and chronic stages of Lyme disease.
  • Osp outer surface proteins
  • Negative Negative values for antibodies to all three Osp antigens are predictive that the horse is not infected. If only one or two values are in the negative range see interpretation for equivocal or positive values for the corresponding Osp antigen.
  • Equivocal Equivocal values can indicate very early infection or can be induced by non-specific serum reactions.
  • the horse should be retested in 2-3 weeks to confirm or exclude early infection. If one or two values are in the positive range see interpretation for positive values forthat corresponding Osp antigen.
  • Positive/OspA >2000: Positive values for antibodies to OspA are typically observed in vaccinated animals. In horses, however, antibodies to OspA also seem to rise during infection. Thus, the interpretation of results on antibodies to OspA is more complex in horses. If antibodies to OspC and/or OspF ate positive, along with OspA, the horse should be considered as infected with B. burgdorferi .
  • Positive/OspC Positive values for antibodies to OspC only ate indicative for early infection. Antibody values for OspA can also be elevated during early infection.
  • Positive/OspF Positive values for antibodies to OspF only are predictive for chronic infection stages. Positive values for antibodies to OspC and OspF in the same sample are indicators for an infection that occurred several weeks ago and is moving towards the chronic stage. Referral test performed at Cornell University. c2 - Interpretation: If your result is: The interpretation is: NEGATIVE No antibody present @ 1:25; POSITIVE @ (titer) Antibody present @ (titer). Positive samples are tested in incremental dilutions to 1:3200.
  • Titers beyond 1:3200 are usually of limited clinical value. If you wish an endpoint titer there is an additional charge. A positive titer indicates exposure to E. canis or similar antigen but does not confirm the presence of disease. A CBC is recommended to identify abnormalities consistent with infection. If confirmation of infection is desired, Ehrlichia PCR test, code 2634 can be useful, especially in clinically sick animals.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Dermatology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Ophthalmology & Optometry (AREA)
  • Molecular Biology (AREA)
  • Physiology (AREA)
  • Emergency Medicine (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Nutrition Science (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurosurgery (AREA)
  • Endocrinology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US16/770,807 2017-12-07 2018-12-07 Compositions and Methods for the Treatment of Metabolic Conditions Abandoned US20200289553A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/770,807 US20200289553A1 (en) 2017-12-07 2018-12-07 Compositions and Methods for the Treatment of Metabolic Conditions

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762595909P 2017-12-07 2017-12-07
US16/770,807 US20200289553A1 (en) 2017-12-07 2018-12-07 Compositions and Methods for the Treatment of Metabolic Conditions
PCT/US2018/064610 WO2019113543A1 (en) 2017-12-07 2018-12-07 Compositions and methods for the treatment of metabolic conditions

Publications (1)

Publication Number Publication Date
US20200289553A1 true US20200289553A1 (en) 2020-09-17

Family

ID=66750649

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/770,807 Abandoned US20200289553A1 (en) 2017-12-07 2018-12-07 Compositions and Methods for the Treatment of Metabolic Conditions
US16/665,795 Active US11344529B2 (en) 2017-12-07 2019-10-28 Compositions and methods for the treatment of metabolic conditions
US17/364,098 Pending US20210322373A1 (en) 2017-12-07 2021-06-30 Compositions and Methods for the Treatment of Metabolic Conditions

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/665,795 Active US11344529B2 (en) 2017-12-07 2019-10-28 Compositions and methods for the treatment of metabolic conditions
US17/364,098 Pending US20210322373A1 (en) 2017-12-07 2021-06-30 Compositions and Methods for the Treatment of Metabolic Conditions

Country Status (17)

Country Link
US (3) US20200289553A1 (ja)
EP (1) EP3720421A4 (ja)
JP (2) JP2021505673A (ja)
KR (1) KR102546138B1 (ja)
CN (1) CN111565709A (ja)
AU (1) AU2018379078A1 (ja)
BR (1) BR112020011417A2 (ja)
CA (1) CA3084369A1 (ja)
CL (1) CL2020001495A1 (ja)
CR (1) CR20200291A (ja)
EA (1) EA202091396A1 (ja)
IL (1) IL275182B2 (ja)
JO (1) JOP20200134A1 (ja)
MA (1) MA51056A (ja)
MX (1) MX2020005879A (ja)
SG (1) SG11202005158PA (ja)
WO (1) WO2019113543A1 (ja)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022183055A1 (en) * 2021-02-26 2022-09-01 Nevakar Injectables Inc. Sodium bicarbonate injectable formulation and methods of use thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11202004540QA (en) 2017-11-17 2020-06-29 Renovion Inc Stable ascorbic acid compositions and methods of using the same
CN116568276A (zh) * 2020-09-30 2023-08-08 雷文制药有限公司 预防和/或治疗炎症的药物组合物和方法
KR102527526B1 (ko) 2020-11-06 2023-05-02 충북대학교 산학협력단 Lstm 기반 구조물의 동적특성 자동추출방법 및 이를 수행하는 시스템
KR102505980B1 (ko) * 2021-11-12 2023-03-06 알고케어 주식회사 사용자의 현재 건강 상태에 최적화된 제형의 조합을 제공하는 디스펜싱 디바이스와 서버 및 그 동작 방법
US11925703B1 (en) 2022-07-29 2024-03-12 Xellia Pharmaceuticals Aps Liquid composition comprising glucose

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150079201A1 (en) * 2008-07-25 2015-03-19 Reven Pharmaceuticals, Inc. Compositions and methods for the prevention and treatment of cardiovascular diseases
CN106924744A (zh) * 2015-12-30 2017-07-07 钟术光 一种供注射或口服用的药物载体(或制剂)

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5080886A (en) * 1990-01-05 1992-01-14 Sterling Drug Inc. Pharmaceutical compositions for the prevention and treatment of oxidant injuries
EP0746202A4 (en) * 1992-01-06 1997-06-25 Health Maintenance Programs COMPOSITION CONTAINING A PHARMACEUTICALLY ACTIVE ANTI-OXIDANT AND METHOD FOR USE IN THE PREVENTION AND TREATMENT OF RESTENOSIS AFTER ANGIOPLASTY
JPH0959150A (ja) * 1995-08-11 1997-03-04 Roussel Morishita Kk 経静脈用輸液製剤
FR2740686B1 (fr) * 1995-11-03 1998-01-16 Sanofi Sa Formulation pharmaceutique lyophilisee stable
US20030190307A1 (en) * 1996-12-24 2003-10-09 Biogen, Inc. Stable liquid interferon formulations
JP2003501488A (ja) 1999-06-11 2003-01-14 ネオルックス コーポレイション 骨髄抑制のための高投与量放射性核種錯体
US20040110684A1 (en) 1999-08-02 2004-06-10 Universite Catholique De Louvain Novel pharmaceutical compositions for modulating angiogenesis
EP1282416A2 (en) * 2000-05-19 2003-02-12 Progenics Pharmaceuticals, Inc. Dehydroascorbic acid formulations and uses thereof
US7122210B2 (en) * 2002-01-11 2006-10-17 Baxter International Inc. Bicarbonate-based solutions for dialysis therapies
WO2005044176A2 (en) 2003-11-10 2005-05-19 Lipogen Ltd. Compositions containing phosphatidic acid, methods of use thereof, methods of manufacture thereof, and articles of manufacture containing same
JP2005179200A (ja) * 2003-12-16 2005-07-07 Terumo Corp ビタミンb1類配合輸液剤
EP1731146A4 (en) * 2004-03-05 2009-07-01 Morishige Fumie PREVENTION AND MEASURE AGAINST MITOCHONDRIA ILLNESS
DE102004023828A1 (de) * 2004-05-13 2005-12-08 Fresenius Medical Care Deutschland Gmbh Lösung für die Peritonealdialyse
CU23432B6 (es) * 2005-11-02 2009-10-16 Ct Ingenieria Genetica Biotech Formulaciones estabilizadas que contienen a los interferones gamma y alfa en proporciones potenciadoras
JP4615470B2 (ja) 2006-03-29 2011-01-19 卓郎 簑和田 大脳の認知力を用いた疾患治療・予防の方法および医薬
US7282225B1 (en) 2006-09-27 2007-10-16 Occular Technologies, Inc. Composition and methods for improving retinal health
CN101209344B (zh) * 2006-12-28 2012-07-04 赵超英 高渗液组合物在制备促进伤口愈合的药物中的应用
SI2805945T1 (sl) 2007-01-10 2019-09-30 Msd Italia S.R.L. Amid substituirani indazoli, kot inhibitorji poli(ADP-riboza)polimeraze(PARP)
RU2513142C2 (ru) * 2008-06-12 2014-04-20 Медтроник Ксомед Инк. Способ лечения хронических ран
EP2456432B1 (en) 2009-07-22 2024-02-28 University of Massachusetts Compositions to reduce oxidative stress
WO2011013138A1 (en) * 2009-07-29 2011-02-03 Strides Arcolab Limited Stable multi- vitamin formulations
MX2012003686A (es) * 2009-10-15 2012-07-25 Purecircle Sdn Bhd Rebaudiosido d de alta pureza y aplicaciones.
EP2324832B2 (de) * 2009-11-09 2016-11-30 Biogena Naturprodukte GmbH & Co KG Nahrungsergänzungsmittel bei Einnahme von hormonellen Kontrazeptiva
CN102869362A (zh) * 2010-05-07 2013-01-09 味之素株式会社 混合有维生素的外周静脉给药用营养输液
US20130288985A1 (en) * 2010-07-15 2013-10-31 The Schepens Eye Research Institute Inc. Compositions and methods of treatment of corneal endothelium disorders
CN103327986B (zh) * 2010-07-22 2018-05-25 雷文制药有限公司 包含使用磁偶极子稳定化溶液的治疗或改善疾病并增强表现的方法
KR20130021958A (ko) * 2011-08-24 2013-03-06 울산대학교 산학협력단 인공 수액
US20150196708A1 (en) * 2012-08-17 2015-07-16 The Regents Of The University Of California Systems, methods and compositions for improved treatment of acidosis
CA2920147C (en) 2013-08-07 2022-09-20 Yeda Research And Development Co. Ltd. Peptides capable of reactivating p53 mutants
EP3419613A4 (en) * 2016-02-23 2019-11-06 Mitronite Inc. COMPOSITIONS AND METHOD FOR IMPROVING MITOCHONDRIAL FUNCTION

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150079201A1 (en) * 2008-07-25 2015-03-19 Reven Pharmaceuticals, Inc. Compositions and methods for the prevention and treatment of cardiovascular diseases
CN106924744A (zh) * 2015-12-30 2017-07-07 钟术光 一种供注射或口服用的药物载体(或制剂)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CN106924744A translation (Year: 2017) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022183055A1 (en) * 2021-02-26 2022-09-01 Nevakar Injectables Inc. Sodium bicarbonate injectable formulation and methods of use thereof

Also Published As

Publication number Publication date
CL2020001495A1 (es) 2021-02-19
CA3084369A1 (en) 2019-06-13
IL275182A (en) 2020-07-30
IL275182B1 (en) 2023-12-01
BR112020011417A2 (pt) 2020-11-24
MA51056A (fr) 2020-10-14
US20210322373A1 (en) 2021-10-21
KR20200097757A (ko) 2020-08-19
SG11202005158PA (en) 2020-06-29
CN111565709A (zh) 2020-08-21
EP3720421A4 (en) 2021-09-15
MX2020005879A (es) 2020-10-07
JP2023134671A (ja) 2023-09-27
IL275182B2 (en) 2024-04-01
US11344529B2 (en) 2022-05-31
WO2019113543A1 (en) 2019-06-13
CR20200291A (es) 2020-11-18
EP3720421A1 (en) 2020-10-14
JOP20200134A1 (ar) 2022-10-30
AU2018379078A1 (en) 2020-07-23
KR102546138B1 (ko) 2023-06-23
JP2021505673A (ja) 2021-02-18
EA202091396A1 (ru) 2020-09-02
US20200121640A1 (en) 2020-04-23

Similar Documents

Publication Publication Date Title
US11344529B2 (en) Compositions and methods for the treatment of metabolic conditions
Canavese et al. Long-term, low-dose, intravenous vitamin C leads to plasma calcium oxalate supersaturation in hemodialysis patients
Racek et al. Influence of chromium-enriched yeast on blood glucose and insulin variables, blood lipids, and markers of oxidative stress in subjects with type 2 diabetes mellitus
Darmaun et al. Parenteral nutrition for preterm infants: Issues and strategy
Teplan et al. Enhanced metabolic effect of erythropoietin and keto acids in CRF patients on low-protein diet: Czech multicenter study
Ditzel et al. Disturbance of inorganic phosphate metabolism in diabetes mellitus: its impact on the development of diabetic late complications
Morgans et al. The role of carnitine in maintenance dialysis therapy
CA3141418A1 (en) Methods and compositions for improving outcomes of cancer patients
Tanner et al. Combined hyperlipidemia in patients with lysinuric protein intolerance
US11357746B2 (en) Method for enhancing energy production and metabolism in cells
US20160095824A1 (en) Use of vitamin k for weight maintenance and weight control
OA19913A (en) Compositions and methods for the treatment of metabolic conditions.
US20100331286A1 (en) Combination therapy for treatment of bone and mineral disorders for patients with impaired renal function
Klein et al. Vitamin and trace element homeostasis following severe burn injury
AU2011343162A1 (en) Use of vitamin K for weight maintenance and weight control
Fassinger et al. Serum retinol, retinol-binding protein, and transthyretin in children receiving dialysis
HamaSalih et al. Effects of vitamin D supplementation on red cell indices and cytokines in patients with thalassemia: An open-label randomized clinical trial
Domrose et al. Vitamins are associated with survival in patients with end-stage renal disease: a 4-year prospective study
CN103417523B (zh) L-谷氨酰胺在制备治疗高同型半胱氨酸血症药物中的应用
Fouque Low protein, amino acid and ketoacid diets to slow the progression of chronic kidney disease and improve metabolic control of uremia
Jover-Ríos et al. Prognostic implications of vitamin D deficiency in chronic kidney disease
Larsen Treatment of congestive heart failure
Cano Noël Nutritional support in renal failure Topic 15
Foody 5 High-Density Lipoprotein Cholesterol, Triglycerides, and Coronary Artery

Legal Events

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: NON FINAL ACTION MAILED

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

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

AS Assignment

Owner name: REVEN IP HOLDCO LLC, COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LANGE, PETER B.;REEL/FRAME:058930/0699

Effective date: 20220203

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

Free format text: FINAL REJECTION MAILED

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

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

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

Free format text: ADVISORY ACTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: REVEN IP HOLDCO LLC, COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REVEN PHARMACEUTICALS, INC.;REEL/FRAME:060708/0472

Effective date: 20210305

Owner name: REVEN, LLC, COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAN WYK, HENDRIK JOHANNES PETRUS;VAN WYK, MARIETTE LUISE;DENOMME, BRIAN DAVID;AND OTHERS;SIGNING DATES FROM 20191106 TO 20191112;REEL/FRAME:060706/0963

Owner name: REVEN PHARMACEUTICALS, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ERVIN, JAMES;VAN WYK, HENDRIK JOHANNES PETRUS;VAN WYK, MARIETTE LUISE;AND OTHERS;SIGNING DATES FROM 20181210 TO 20191220;REEL/FRAME:060706/0819

Owner name: REVEN IP HOLDCO LLC, COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REVEN, LLC;REEL/FRAME:060708/0295

Effective date: 20200605

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

Free format text: FINAL REJECTION MAILED

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

Free format text: ADVISORY ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION