US20220354812A1 - Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy - Google Patents

Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy Download PDF

Info

Publication number
US20220354812A1
US20220354812A1 US17/552,660 US202117552660A US2022354812A1 US 20220354812 A1 US20220354812 A1 US 20220354812A1 US 202117552660 A US202117552660 A US 202117552660A US 2022354812 A1 US2022354812 A1 US 2022354812A1
Authority
US
United States
Prior art keywords
composition
subject
cancer
cysteine
vitamin
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
US17/552,660
Inventor
Xiyan Li
Xin Wang
Xiaolu Yang
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.)
Filtricine Inc
Original Assignee
Filtricine Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Filtricine Inc filed Critical Filtricine Inc
Priority to US17/552,660 priority Critical patent/US20220354812A1/en
Assigned to FILTRICINE, INC. reassignment FILTRICINE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LI, Xiyan, WANG, XIN, YANG, XIAOLU
Publication of US20220354812A1 publication Critical patent/US20220354812A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/28Peritoneal dialysis ; Other peritoneal treatment, e.g. oxygenation
    • A61M1/287Dialysates therefor
    • 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
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/15Vitamins
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/16Inorganic salts, minerals or trace elements
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/17Amino acids, peptides or proteins
    • A23L33/175Amino acids
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/30Dietetic or nutritional methods, e.g. for losing weight
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/07Retinol compounds, e.g. vitamin A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/14Quaternary ammonium compounds, e.g. edrophonium, choline
    • 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
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • A61K31/355Tocopherols, e.g. vitamin E
    • 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
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • 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
    • 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
    • A61K31/59Compounds containing 9, 10- seco- cyclopenta[a]hydrophenanthrene ring systems
    • A61K31/5939,10-Secocholestane derivatives, e.g. cholecalciferol, i.e. vitamin D3
    • 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
    • A61K31/7004Monosaccharides having only carbon, hydrogen and oxygen atoms
    • 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
    • A61K33/04Sulfur, selenium or tellurium; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/16Fluorine compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/18Iodine; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/26Iron; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/30Zinc; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/32Manganese; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/34Copper; Compounds thereof
    • 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
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis
    • A61M1/16Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis with membranes
    • A61M1/1654Dialysates therefor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/05General characteristics of the apparatus combined with other kinds of therapy

Definitions

  • Cancer and obesity are major public health problems. Cancer is the second leading cause of death globally, and was responsible for 8.8 million deaths in 2012. Globally, approximately 1 in 6 deaths is due to cancer. Cancer arises from the transformation of normal cells into tumor cells in a multistage process that generally progresses from a pre-cancerous lesion to a malignant tumor.
  • One defining feature of cancer is the rapid creation of abnormal cells that grow beyond their usual boundaries, and which can then invade adjoining parts of the body and spread to other organs.
  • Cancer cells adopt a distinctive type of metabolism, the “Warburg effect,” to promote growth, survival, proliferation, and long-term maintenance. This altered metabolism involves uncoupled cellular glycolysis and mitochondrial aerobic respiration, which may lead to the requirement of certain nutrients that are not necessary for normal cells.
  • Obesity is a medical condition in which excess body fat has accumulated to the extent that it may have a negative impact on health. This condition is commonly caused by a combination of excessive food intake, lack of physical activity, and genetic susceptibility. Obesity increases the likelihood of various diseases and conditions, such as, cardiovascular diseases, type 2 diabetes, obstructive sleep apnea, cancer, osteoarthritis and depression.
  • the present disclosure relates to providing a subject with a treatment that includes the control of a subject's nutritional diet and/or dialysis.
  • treating a patient in need thereof may comprise placing the patient on a cysteine depleted diet and placing the patient under dialysis.
  • the present disclosure provides solid and liquid compositions that are depleted with one or more nutrients, such as nonessential amino acids, and methods of treating cancer, renal disease, and metabolic diseases, such as obesity, with the compositions.
  • the present disclosure provides for facilities, systems and kits that relate to the distribution and/or performance of the described compositions and methods.
  • the present disclosure utilizes hemodialysis and peritoneal dialysis as a way to deplete one or more nutrients in the subject by administering compositions that are lacking in such nutrients.
  • Hemodialysis involves withdrawing blood from the body and cleaning it in an extracorporeal blood circuit and then returning the cleansed blood to the body.
  • the extracorporeal blood circuit includes a dialyzer which comprises a semipermeable membrane. Within this dialyzer waste substances and excess fluid is removed through the semipermeable membrane, and the semipermeable membrane within the dialyzer has a blood side and a dialysate side.
  • Peritoneal dialysis involves the use of the peritoneum in a person's abdomen as the membrane through which fluid and dissolved substances are exchanged with the blood.
  • Peritoneal dialysis is used to remove excess fluid, correct electrolyte problems, and remove toxins in those with kidney failure.
  • peritoneal dialysis a solution is introduced through a permanent tube in the lower abdomen and then removed. This may either occur at regular intervals throughout the day, known as continuous ambulatory dialysis, or at night with the assistance of a machine, known as automated peritoneal dialysis.
  • the present disclosure envisions utilizing hemodialysis and peritoneal dialysis as way to provide a subject in need thereof with a particular nutritional makeup by depleting and/or restoring one or more nutrients in the subject.
  • the present disclosure provides dialysis systems and equipment thereof.
  • a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides a liquid composition
  • a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides a liquid composition
  • a liquid composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (c) at least one essential amino acid; and (d) an aqueous solution, wherein the liquid composition comprises at least 10 ⁇ M of essential amino acid, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein the liquid composition has a pH of 4.0-6.5, and wherein the liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the carbohydrate is selected from a group consisting of dextrose and glucose.
  • the carbohydrate may be dextrose.
  • the amount of dextrose is 1.0-4.5% w/v.
  • the amount of dextrose may be 1.5-4.25% w/v.
  • the amount of dextrose may be 1.7-4.0% w/v.
  • the amount of dextrose is 0.05-0.50% w/v.
  • the amount of dextrose may be 0.05-0.25% w/v.
  • the amount of dextrose may be 0.08-0.22% w/v.
  • the amount of dextrose may be 0.1-0.2% w/v.
  • the liquid composition comprises at least 10 ⁇ M of said essential amino acid.
  • the amount of at least one of the essential amino acid is 10-300 ⁇ M.
  • the amount of at least one of the essential amino acid is 20-250 ⁇ M.
  • the essential amino acid may be selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine.
  • at least one of the essential amino acid is methionine.
  • at least one of the essential amino acid is arginine.
  • the liquid composition may comprise at least two essential amino acids.
  • the liquid composition may comprise at least three essential amino acids.
  • the liquid composition may comprise at least four essential amino acids.
  • the liquid composition may comprise at least five essential amino acids.
  • the liquid composition may comprise at least six essential amino acids.
  • the liquid composition may comprise at least seven essential amino acids.
  • the liquid composition may comprise at least eight essential amino acids.
  • the liquid composition may comprise at least nine essential amino acids.
  • the liquid composition may comprise at least ten essential amino acids.
  • the liquid composition may further comprise at least one vitamin.
  • the vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline, such as for example, the vitamin is vitamin C.
  • the amount of the vitamin is about 0.000010 to 200 ⁇ M.
  • the liquid composition comprises at least 1 ⁇ M of vitamin C.
  • the liquid composition may comprise at least 7 ⁇ M of vitamin C.
  • the vitamin is vitamin B.
  • the liquid composition may comprise 0.000003 to 1500 ⁇ M of said vitamin B.
  • the liquid composition may further comprise an essential fatty acid.
  • the essential amino acid may be an omega-3 fatty acid.
  • the amount of glutamate is approximately 2-500 ⁇ M.
  • the amount of glutamate may increase glutamate in the blood by at least 150 ⁇ M.
  • the liquid composition further comprises at least one mineral.
  • the mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc.
  • the liquid composition may comprise at least 5 nM of the mineral. In some embodiments, the amount of at least one mineral is 5 nM-5 ⁇ M.
  • the liquid composition is stored in a container.
  • the container may be a dialysis bag.
  • the dialysis bag may be intended for single-use.
  • the container holds 1500 to 5000 mL of the liquid composition. For example, approximately 2000 mL of the liquid composition is a single unit. In another embodiment, approximately 2500 mL of the liquid composition is a single unit.
  • the amount of at least one ion is at least approximately 0.1 mEq/L.
  • the amount of at least one ion may be 0.1-150 mEq/L.
  • liquid composition is free of nonessential amino acids other than glutamate.
  • the liquid composition may further comprise at least one nonessential amino acid.
  • the amount of the nonessential amino acid is at least approximately 5 ⁇ M.
  • the amount of the nonessential amino acid may be 5-600 ⁇ M.
  • the nonessential amino acid may be selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, and tyrosine.
  • the liquid composition comprises 0-4 ⁇ M of cysteine and/or cystine.
  • the liquid composition may comprise 0-1 ⁇ M of cysteine and/or cystine.
  • the liquid composition has a pH of approximately 5.0-6.5.
  • the liquid composition may have a pH of approximately 5.5-6.0.
  • the liquid composition may have a pH of approximately 6.0.
  • the liquid composition may have a pH of approximately 5.5.
  • the liquid composition may have a pH of approximately 5.0.
  • the liquid composition has an osmolarity of 300-500.
  • the liquid composition may have an osmolarity of 300-400.
  • the liquid composition may have an osmolarity of 400-500.
  • the liquid composition may have an osmolarity of 300-350.
  • the aqueous solution is sterile. In another embodiment, the aqueous solution is nonsterile. In some embodiments, the liquid composition is a dialysis solution.
  • the present disclosure provides, a dry composition
  • a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the dry composition optionally comprises glutamate, wherein the dry composition comprises 0-5 ⁇ M of cysteine and/or cystine.
  • the present disclosure provides a dry composition
  • a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the dry composition optionally comprises glutamate, wherein the dry composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, and wherein the dry composition comprises 0-5 ⁇ M of cysteine and/or cystine.
  • the carbohydrate is selected from a group consisting of dextrose, glucose, sucrose, fructose, and lactose.
  • the carbohydrate may be dextrose.
  • the present disclosure provides a dry composition
  • a dry composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (c) at least one essential amino acid, wherein the dry composition comprises at least 2 ⁇ g of essential amino acid, and wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • the amount of dextrose is at least approximately 10 g.
  • the amount of dextrose may be 10-50 g, such as approximately 50 g.
  • the amount of dextrose is at least approximately 0.5 g.
  • the amount of dextrose may be 0.5-2.5 g, such as approximately 2 g.
  • the essential amino acid is selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine.
  • at least one essential amino acid is methionine.
  • at least one essential amino acid is arginine.
  • the dry composition comprises at least two essential amino acids.
  • the dry composition may comprise at least four essential amino acids.
  • the dry composition may comprise at least five essential amino acids.
  • the dry composition may comprise at least six essential amino acids.
  • the dry composition may comprise at least seven essential amino acids.
  • the dry composition may comprise at least eight essential amino acids.
  • the dry composition may comprise at least nine essential amino acids.
  • the dry composition may comprise at least ten essential amino acids.
  • the dry composition comprises at least 2 ⁇ g of the essential amino acid.
  • the dry composition may comprise at least one vitamin.
  • the vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline.
  • the amount of the vitamin is about 0.01-1500 ⁇ m.
  • the vitamin is vitamin C.
  • the dry composition may comprise at least 1000 ⁇ g of vitamin C.
  • the dry composition comprises at least 2500 ⁇ g of vitamin C.
  • the dry composition comprises at least 0.01 ⁇ g of said vitamin B.
  • the dry composition may comprise at least 1 ⁇ g of vitamin B.
  • the amount of said glutamate is approximately 0.1 mg to 50 mg.
  • the liquid composition may be free of nonessential amino acids other than glutamate.
  • the dry composition may further comprise an essential fatty acid, such as an omega-3 fatty acid.
  • the dry composition may further comprise at least one mineral.
  • the mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc.
  • the dry composition comprises at least 0.3 ⁇ g of mineral.
  • the dry composition may comprise at least 1 ⁇ g of mineral.
  • the dry composition is stored in a container.
  • the container may be a dialysis bag.
  • the dialysis bag may be intended for single-use.
  • the dry composition may be a dialysis mixture.
  • the dry composition further comprises an aqueous solution in the container, thereby producing a mixture.
  • the container may comprise approximately 2000 mL of aqueous solution.
  • the container may comprise approximately 2500 mL of aqueous solution.
  • the mixture may have a pH of 4.0-6.5.
  • the mixture may have an osmolarity of 300-720 mOsmol/L.
  • the amount of at least one ion is at least 100 ⁇ mol.
  • the dry composition further comprises at least one nonessential amino acid.
  • the amount of nonessential amino acid may be at least 1 ⁇ g. In some embodiments, the amount of nonessential amino acid is 1-100 ⁇ g.
  • the dry composition may comprise 0-5 mg of cysteine and/or cystine. In some embodiments, the dry composition comprises 0-1 mg of cysteine and/or cystine.
  • the present disclosure provides a concentrated composition comprising a dry composition described herein, and an initial aqueous solution, wherein the dry composition is dissolved in the initial aqueous solution.
  • the present disclosure provides a food composition
  • a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least one nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline, wherein each nonessential amino acid is 0-1% wt/wt of protein in the food composition, wherein the food composition comprises 0-1% cysteine and/or cystine wt/wt of protein in the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • the amount of nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline is 0-0.1% wt/wt of protein in the food composition.
  • the food composition comprises at least two nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least three nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least four nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least five nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least six nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the nonessential amino acid is glycine.
  • the nonessential amino acid is proline.
  • the amount of cysteine is 0-0.1% wt/wt of protein in the food composition.
  • the present disclosure provides a food composition
  • a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least six nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine, wherein each nonessential amino acid is 0-1% wt/wt of protein in the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • the amount of each nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine is 0-50%.
  • the food composition comprises at least seven nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least eight nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least nine of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least ten nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • at least six nonessential amino acids are selected from the group consisting of glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • at least one nonessential acid is cysteine.
  • the amount of cysteine and/or cystine may be 0-0.04% wt/wt of protein in the food composition.
  • the present disclosure provides a food composition comprising: (a) at least one essential amino acid; and (b) glutamate, wherein the food composition is substantially free of nonessential amino acids other than glutamate.
  • the amount of the cysteine devoid protein source is at least 1 wt/wt of the food composition.
  • the amount of the cysteine devoid protein source may be 1-5% of the food composition.
  • the cysteine devoid protein source is gelatin.
  • the food composition comprises approximately 0.1 mg to 50 mg of glutamate.
  • the essential amino acid is selected from a group consisting of arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.
  • the essential amino acid is methionine.
  • the essential amino acid may be at least 1.0% wt/wt of the food composition.
  • the amount of essential amino acid may be 2.0-75% wt/wt of the food composition.
  • the food composition comprises at least two essential amino acids.
  • the food composition may comprise at least three essential amino acids.
  • the food composition may comprise at least four essential amino acids.
  • the food composition may comprise at least five essential amino acids.
  • the food composition may comprise at least six essential amino acids.
  • the food composition may comprise at least seven essential amino acids.
  • the food composition may comprise at least eight essential amino acids.
  • any food composition described herein further comprises a pharmaceutically active agent.
  • the pharmaceutically active agent may be an anti-cancer therapeutic.
  • the food composition is packaged as a solid food, semi-solid food, or beverage.
  • the food composition is packaged as a food product selected from a group consisting of a snack bar, cereal product, bakery product, and dairy product.
  • the food composition may be baby formula.
  • the food composition may be a soft drink.
  • the food composition may be a nutritional shake.
  • the food composition is a granola bar.
  • the food composition comprises 10-20% w/w of total amino acids.
  • the food composition may comprise approximately 17% w/w of total amino acids.
  • the present disclosure provides a facility comprising a plurality of the liquid compositions described herein.
  • the facility may be a pharmaceutical manufacturing site.
  • the facility may be a distribution center.
  • the facility may be a hospital or clinic.
  • the present disclosure provides a plurality of the dry compositions described herein.
  • the facility may be a pharmaceutical manufacturing site.
  • the facility may be a distribution center.
  • the facility may be a hospital or clinic.
  • the present disclosure provides a plurality of the food composition described herein.
  • the facility may be a food manufacturing site.
  • the facility may be a distribution center.
  • the facility may be a commercial kitchen, restaurant or eating facility.
  • the present disclosure provides a kit comprising: (a) a pre-packaged liquid composition described herein; and (b) instructions for using the pre-packaged liquid composition.
  • the kit provides complete treatments for at least one week.
  • the kit may provide complete treatments for at least four weeks.
  • the present disclosure provides a kit comprising: (a) a pre-packaged dry composition or concentrated composition described herein; and (b) instructions for using the pre-packaged dry composition or concentrated composition.
  • the kit provides complete treatments for at least one week. For example, the kit provides complete treatments for at least four weeks.
  • the present disclosure provides a kit comprising: (a) a pre-packaged food composition described herein; and (b) instructions for using the pre-packaged food composition.
  • the kit provides complete meals for at least one day.
  • the kit may provide complete meals for at least five days.
  • the kit further comprises a pre-packaged liquid composition described herein.
  • the kit may further comprise instructions for using the pre-packaged liquid composition.
  • the kit further comprises a pre-packaged dry composition described herein.
  • the kit further comprises instructions for using the pre-packaged dry composition.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • the cancer may be selected from the group consisting of liver cancer, breast cancer, lung cancer, leukemia, prostate, and colon cancer.
  • the subject may be human.
  • the subject may be an animal.
  • the subject has a genetic predisposition for cancer.
  • the subject may have elevated cysteine biosynthetic gene expression levels.
  • treating cancer is characterized by a reduction in tumor size.
  • the tumor may be reduced by at least 30%.
  • the tumor may be reduced by at least 50%.
  • the tumor may be reduced by at least 80%.
  • treating cancer is characterized by decreased levels of plasma cysteine.
  • the levels of plasma cysteine may be reduced by at least 30%.
  • the levels of plasma cysteine may be reduced by at least 50%.
  • the levels of plasma cysteine may be reduced by at least 80%.
  • the levels of plasma cysteine may be reduced by at least 90%.
  • the levels of plasma cysteine are reduced by at least 95%.
  • the present disclosure provides a method for reducing tumor size in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • the present disclosure provides a method for reducing tumor size in a subject in need thereof, the method comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • the present disclosure provides a method for reducing tumor size in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • the subject may be human.
  • the subject may be an animal.
  • the subject has a genetic predisposition for cancer.
  • the subject may have elevated cysteine biosynthetic gene expression levels.
  • reducing tumor size is characterized by a reduction in tumor size.
  • the tumor may be reduced by at least 30%.
  • the tumor may be reduced by at least 50%.
  • the tumor may be reduced by at least 80%.
  • the present disclosure provides a method for treating obesity, the method comprising administering to the subject any food composition described herein.
  • the subject may be human.
  • the subject may be an animal.
  • the subject has a genetic predisposition for obesity.
  • treating obesity is characterized by the subject losing at least 10% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 20% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 30% of the subject's initial body weight.
  • the present disclosure provides a method for treating renal disease, the method comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • the subject may be human.
  • the subject may be an animal.
  • the subject is diagnosed with chronic kidney disease.
  • the methods further comprise utilizing a membrane.
  • the membrane comprises modified cellulose diacetate or polysulfone.
  • the membrane is a low-flux membrane.
  • the low-flux membrane may permit small molecule exchange.
  • the low-flux membrane comprises a plurality of pores, and the plurality of pores comprises a first end and a second end. The plurality of pores may have an average pore diameter of approximately 1.8 nm.
  • the low-flux membrane permits approximately 50% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end.
  • the low-flux membrane may permit approximately 75% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end.
  • the low-flux membrane may permit approximately 90% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end.
  • the low-flux membrane may permit approximately 95% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end.
  • approximately 20% of a molecule of at least 1000 Daltons passes through the plurality of pores, from the first end to the second end. For example, approximately 10% of a molecule of at least 1000 Daltons may pass from the first side to the second side. Approximately 1% of a molecule of at least 1000 Daltons may pass from the first side to the second side.
  • the composition is administered to the subject at least once a week.
  • the composition may be administered to the subject at least three times a week.
  • the composition may be administered to the subject at least five times a week.
  • the liquid composition, concentrated composition, or dry composition is administered to the subject over the course of at least one week.
  • the composition may be administered to the subject over the course of at least four weeks.
  • the composition may be administered to the subject over the course of at least 2 months. In an exemplary embodiment, the composition is administered to the subject over the course of at least 3 months.
  • the food composition is administered to the subject at least once a week.
  • the food composition may be administered to the subject at least three times a week.
  • the food composition may be administered to the subject at least five times a week.
  • the food composition may be administered to the subject at least twenty-one times a week.
  • the food composition is administered to the subject over the course of at least one week.
  • the food composition may be administered to the subject over the course of at least four weeks.
  • the food composition may be administered to the subject over the course of at least 2 months.
  • the food composition may be administered to the subject over the course of at least 3 months.
  • the compositions may be administered to the subject sequentially. In another embodiment, the compositions may be administered to the subject simultaneously. In some embodiments, the compositions are administered to the subject at least once a week. For example, the compositions may be administered to the subject at least three times a week. In some embodiments, the compositions are administered to the subject over the course of at least one week. For example, the compositions may be administered to the subject over the course of at least four weeks.
  • the methods further comprise administering a pharmaceutically active agent.
  • the pharmaceutically active agent may be an anti-cancer therapeutic.
  • the pharmaceutically active agent may be provided in a subtherapeutic amount.
  • the methods further comprise administering a nutritional supplement.
  • the nutritional supplement may comprise at least one amino acid selected from a group consisting of arginine, histidine, isoleucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.
  • the nutritional supplement provides the subject with nutrients at levels in accordance with the Recommended Dietary Allowances (RDAs), Adequate Intakes (AIs), and/or Daily Value (DV).
  • RDAs Recommended Dietary Allowances
  • AIs Adequate Intakes
  • DV Daily Value
  • the disclosure provides a method of treating cancer in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • the disclosure provides a method of depleting cysteine in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • the disclosure provides a method of treating cancer in a subject that has been placed on a dialysis procedure to reduce cysteine in the blood, comprising: placing the subject on a low-cysteine diet.
  • the dialysis comprises administering a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein.
  • the low-cysteine diet is a food composition disclosed herein.
  • the disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein.
  • the disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein, and a dialysis machine configured to administer the liquid composition, the concentrated composition or the dry composition.
  • the dialysis machine comprises a pump, a pressure monitor, air trap, air detector, a clamp, and/or a filter.
  • FIG. 1 depicts a metabolic cancer therapy in which a cancer patient is treated with dialysis and/or a nutritional diet in order to deplete targeted nutrient(s).
  • FIG. 2 shows an overview of the diets used in colon, breast, prostate, and pancreatic cancer studies.
  • FIG. 3 shows an overview of the diet compositions used in the studies of the present disclosure.
  • the mice used for the study were normal and active (other than tumor-bearing.)
  • FIG. 4 depicts a graph showing the effects of a complete diet and MEAD202 on tumor volume (mm 3 ) in colon cancer (HCT116). Mice with implanted HCT116 cells were on diet treatment on day 7. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 5A depicts a graph showing the effects of a complete diet (Ctrl) and treatment diets on tumor volume (mm 3 ) over time (days after tumor implant) in triple negative breast cancer (MDA-MDB-231). Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 5B depicts a graph showing the effects of a complete diet (Ctrl), MEAD202 and MEAD203 on tumor volume (mm 3 ) over time (days after tumor inoculation) in triple negative breast cancer (MDA-MDB-231). Mice with implanted MDA-MDB-231 cells were on diet treatment on day 7. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 6 depicts a graph showing the effects of a complete diet (Ctrl) and MEAD203 on tumor volume (mm 3 ) over time (days after tumor inoculation) in prostate cancer (PC3). Mice with implanted PC-3 cells were on diet treatment on day 5. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 7 depicts a graph showing the effects of a complete diet (Ctrl) and treatment diets on tumor volume (mm 3 ) over time (days after treatment) in pancreatic cancer (MIAPaCa-2). Mice with implanted MIAPaCa-2 cells were on diet treatment on day 0. The tumor sizes were monitored 2 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 8 shows an overview of the dialysis compositions used in the studies of the present disclosure.
  • FIG. 9 depicts a graph showing the effects of a complete diet (Ctrl), MEAD203+dialysis, MEAD203 on tumor volume (mm 3 ) over time (days after tumor inoculation) in glioblastoma (U87MG, single mouse study). Mice with implanted U87MG cells were on diet treatment on day 5. The tumor sizes were monitored daily. Each treatment group had 1 mouse.
  • the dialysis fluid was MEAD101 (Ctrl) at day 7, 8, 9, 13, 14, 28, 37, and 40.
  • the dialysis fluid was MEAD102 at day 23, 26, 27, 30, 31, 33, 34, 35, 36, 41, and 42.
  • the dialysis fluid was MEAD103 at 43, 44, 47, 48, 49, 50, 51, 54, 55, 56, 57 and 58.
  • FIG. 10 depicts a graph showing the effects of a complete diet, diet only, dialysis only, and diet+dialysis on tumor volume (mm 3 ) over time (days after treatment) in glioblastoma (U87MG). Mice with implanted U87MG cells were on diet treatment on day 1. The tumor sizes were monitored daily. Each group had 6 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 11A depicts a graph showing the effects of a complete diet (Ctrl), MEAD203, MEAD202, and natural chow on body weight (%) over time (days after tumor inoculation) in triple negative breast cancer (MDA-MB-231).
  • Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 11B depicts a graph showing the effects of a complete diet (Ctrl), and MEAD203 on body weight (%) over time (days after tumor inoculation) in prostate cancer (PC3). Mice with implanted PC-3 cells were on diet treatment on day 5. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001
  • FIG. 12A depicts a graph showing the effects of natural chow, complete diet, MEAD202, MEAD203 on glucose (mg/dL) in triple negative breast cancer (MDA-MB-231). Glucose in plasma was measured with Nova Max Glucose Test Strips. Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 12B depicts a graph showing the effects of complete diet and MEAD203 on glucose (mg/dL) in prostate cancer (PC3).
  • Glucose in plasma was measured with Nova Max Glucose Test Strips. Mice with implanted PC-3 cells were on diet treatment on day 5. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 13A depicts a graph showing the effects of natural chow, complete diet, MEAD202, MEAD203 on ⁇ -Ketone (mmol/L) in triple negative breast cancer (MDA-MB-231). Beta-ketone body in plasma was measured with Nova Max Ketone Test Strips. Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 13B depicts a graph showing the effects of complete diet and MEAD203 on ⁇ -Ketone (mmol/L) in prostate cancer. Beta-ketone body in plasma was measured with Nova Max Ketone Test Strips. Mice with implanted PC-3 cells were on diet treatment on day 5. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 14A and FIG. 14B show exemplary dialysis systems, CAPD and CCPD respectively, that is configured to use administer the compositions described herein.
  • FIG. 15 shows and exemplary dialysis system for hemodialysis that is configured to use administer the compositions described herein.
  • FIG. 16 depicts the typical hemodialysis process.
  • any figure or number or amount presented herein is approximate, and that any numerical range includes the minimum number and the maximum number defining the range, whether the word “inclusive” or the like is employed or not, unless implicitly or explicitly understood or stated otherwise.
  • the term “approximately” or “about” or the symbol “—” in reference to a figure or number or amount includes numbers that fall within a range of ⁇ 5% of same, unless implicitly or explicitly understood or stated otherwise.
  • any heading employed is by way of convenience, not by way of limitation.
  • any permissive, open, or open-ended language encompasses any relatively permissive to restrictive language, less open to closed language, or less open-ended to closed-ended language, respectively, unless implicitly or explicitly understood or stated otherwise.
  • the word “comprising” may encompass “comprising”, “consisting essentially of”, and/or “consisting of” type language.
  • administer are defined as the providing a composition to a subject via a route known in the art, including but not limited to intravenous, intraarterial, oral, parenteral, buccal, topical, transdermal, rectal, intramuscular, subcutaneous, intraosseous, transmucosal, or intraperitoneal routes of administration.
  • oral routes of administering a composition may be preferred.
  • administering a composition through dialysis may be preferred.
  • agent refers to a biological, pharmaceutical, or chemical compound or other moiety.
  • Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, a peptide nucleic acid (PNA), an oligonucleotide (including e.g., aptamer and polynucleotides), an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, a branched chain amino acid in free amino acid form or metabolite thereof, or a chemotherapeutic compound.
  • PNA peptide nucleic acid
  • oligonucleotide including e.g., aptamer and polynucleotides
  • an antibody an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, a branched chain amino acid in free amino acid form or metabolite thereof, or a chemotherapeutic compound.
  • protein includes an amino acid mixture comprising
  • oligomers e.g., oligopeptides and oligonucleotides
  • synthetic organic compounds based on various core structures.
  • natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • co-administration can encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the subject at the same time.
  • Co-administration can encompass treatment regimens in which the composition and additional therapeutic agent are administered by the same or different route of administration or at the same or different times.
  • Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
  • Co-administration can include simultaneous administration of the agents in separate compositions, administration at different times in separate compositions, and/or administration in a single composition comprising each of the agents to be co-administered.
  • determining means, “measuring”, “evaluating”, “assessing,” “assaying,” “interrogating,” and “analyzing” are used interchangeably herein to refer to any form of measurement, and include determining if an element is present or not. These terms include both quantitative and/or qualitative determinations. Assessing may be relative or absolute.
  • the term “effective amount” or “therapeutically effective amount” refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below.
  • the therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or down regulation of activity of a target protein.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • essential amino acid or “indispensable amino acid” refers to an amino acid that cannot be synthesized de novo by the organism.
  • Essential amino acids include arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.
  • Essential amino acids may include salts thereof.
  • free refers to compositions that have about 0% of a specific component.
  • nonessential amino acid or “dispensable amino acid” refers to an amino acid that can be synthesized de novo by the organism. Nonessential amino acids include alanine, asparagine, aspartate, cysteine (or cystine), glutamic acid, glutamine, glycine, proline, serine, and tyrosine.
  • cysteine or cystine
  • glutamic acid glutamine
  • glutamine glycine
  • proline proline
  • serine serine
  • prevention refers to providing treatment prior to the onset of a condition. If treatment is commenced in subjects with a condition, such treatment is expected to prevent, or to prevent the progression of, the medical sequelae of the condition.
  • the term “subject” or “individual” includes mammals.
  • Non-limiting examples of mammals include humans and animals, such as mice, including transgenic and non-transgenic mice.
  • the methods described herein can be useful in both human therapeutics, pre-clinical, and veterinary applications.
  • the subject is an animal, and in some embodiments, the subject is human.
  • Other mammals include, and are not limited to, apes, chimpanzees, orangutans, monkeys; domesticated animals (pets) such as dogs, cats, guinea pigs, hamsters, mice, rats, rabbits, and ferrets; domesticated farm animals such as cows, buffalo, bison, horses, donkey, swine, sheep, and goats; or exotic animals typically found in zoos, such as bear, lions, tigers, panthers, elephants, hippopotamus, rhinoceros, giraffes, antelopes, sloth, gazelles, zebras, wildebeests, prairie dogs, koala bears, kangaroo, pandas, giant pandas, hyena, seals, sea lions, and elephant seals.
  • domesticated animals such as dogs, cats, guinea pigs, hamsters, mice, rats, rabbits, and ferrets
  • domesticated farm animals
  • compositions that have less than about 10%, less than about 5%, less than about 1%, less than about 0.5%, less than 0.1% or even less of a specified component.
  • a “subtherapeutic amount” or “sub-therapeutic amount” of an agent, an activator or a therapy is an amount less than the effective amount for that agent, activator or therapy, but when combined with an effective or sub-therapeutic amount of another agent or therapy can produce a desired result, due to, for example, synergy in the resulting efficacious effects, and/or reduced side effects.
  • a sub-therapeutic amount of the agent or component can be such that it is an amount below which would be considered therapeutic.
  • FDA guidelines can suggest a specified level of dosing to treat a particular condition, and a sub-therapeutic amount would be any level that is below the FDA suggested dosing level.
  • the sub-therapeutic amount can be about 1, 5, 10, 15, 20, 25, 30, 35, 50, 75, 90, or 95% less than the amount that is considered to be a therapeutic amount.
  • the therapeutic amount can be assessed for individual subjects, or for groups of subjects.
  • the group of subjects can be all potential subjects, or subjects having a particular characteristic such as age, weight, race, gender, or physical activity level.
  • composition refers to a dry composition, a concentrated composition, a liquid composition, and/or a food composition.
  • compositions such as dry compositions, concentrated compositions, liquid compositions, and food compositions, effective for treating cancer, other metabolic diseases, including obesity, and renal disease.
  • the compositions can be depleted or reduced with respect to one or more amino acids or other nutrients and/or supplemented with respect to one or more other amino acids or nutrients.
  • the depleted or reduced nutrients can be certain nutrients acids that are demanded at a higher degree by cancerous cells as compared to non-cancerous cells.
  • cancerous cells may have a higher nutritional or metabolic requirement for certain non-essential amino acids as compared to non-cancerous cells and so a composition may formulated to be depleted or reduced with respect to those non-essential amino acids while maintaining the normal or supplemented amounts of other nutrients.
  • the compositions may be a composition for dialysis or for a nutritional diet.
  • a dry composition, a concentrated composition, or a liquid composition may also be referred as a “dialysate composition,” “dialysate,” a “dialysis solution,” or a “dialysis composition.
  • a food composition may also be referred to as a “nutritional diet” or a “diet.”
  • the present disclosure provides dialysis compositions.
  • the dialysis composition may be in the form of a liquid composition.
  • the present disclosure provides a liquid composition comprising glutamate.
  • the liquid composition comprises glutamate and at least one essential amino acid.
  • the present disclosure also provides a dialysis composition comprising at least one vitamin.
  • the liquid composition comprises at least one vitamin and at least one essential amino acid.
  • the liquid composition is substantially free of cysteine and/or cystine.
  • a liquid composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one vitamin; (d) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (e) at least one essential amino acid; and (f) an aqueous solution, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides, a liquid composition
  • a liquid composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition is substantially free of nonessential amino acids other than glutamate, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides a liquid composition
  • a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, wherein the liquid composition comprises 0-5 ⁇ M of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides a liquid composition
  • a liquid composition comprising: (a) carbohydrate; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (c) at least one essential amino acid; and an aqueous solution, wherein the liquid composition comprises 0-50 ⁇ M of cysteine and/or cystine, wherein liquid composition has a pH of 4.0-6.5, and wherein the liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides a liquid composition
  • a liquid composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (c) at least one essential amino acid; and an aqueous solution, wherein the liquid composition comprises at least 10 ⁇ M of essential amino acid, wherein the liquid composition comprises 0-50 ⁇ M of cysteine and/or cystine, wherein liquid composition has a pH of 4.0-6.5, and wherein the liquid composition has an osmolarity of 300-720 mOsmol/L.
  • the present disclosure provides a liquid composition
  • a liquid composition comprising: (a) carbohydrate; (b) at least one essential amino acid; and an aqueous solution, wherein the liquid composition comprises 0-50 ⁇ M of cysteine and/or cystine, and wherein the liquid composition is substantially free of nonessential amino acids.
  • the carbohydrate is selected from a group consisting of dextrose, glucose, sucrose, fructose, and lactose.
  • the carbohydrate is dextrose.
  • the liquid composition disclosed herein may comprise amount of dextrose that is 0.1-4.5% w/v.
  • the amount of the dextrose is 1.0-4.5% w/v.
  • the amount of the dextrose may be 1.5-4.25% w/v.
  • the amount of the dextrose is 1.7-4.0% w/v.
  • the amount of dextrose is 0.05-0.25% w/v.
  • the amount of the dextrose may be 0.08-0.22% w/v.
  • the amount of the dextrose is 0.1-0.2% w/v.
  • the amount of dextrose in the liquid composition may be at least, approximately, or no more than 0.05, 0.1, 0.5, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, or 5.0% w/v.
  • the amount of dextrose in the liquid composition may depend on its intended application, such as use in peritoneal dialysis (PD) or use in hemodialysis (HD).
  • the amount of dextrose in the liquid composition may be 1.0-4.5% w/v, such as 1.5-4.25% w/v or 1.7-4.0% w/v.
  • the amount of dextrose in the liquid composition may be 0.05-0.50% w/v, such as 0.08-0.25% w/v or 0.1-0.25% w/v.
  • the liquid composition comprises at least one essential amino acid selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine.
  • at least one of the essential amino acids is methionine.
  • at least one of the essential amino acids is arginine.
  • the amount of at least one of the essential amino acid is at least 10 ⁇ M.
  • the amount of at least one of the essential amino acid may be 10-300 ⁇ M.
  • the amount of at least one of the essential amino acid may be 20-250 ⁇ M.
  • the amount of at least one of the essential amino acid may be at least, approximately, or no more than 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, or 300 ⁇ M.
  • the liquid composition comprises at least two essential amino acids.
  • the liquid composition may comprise at least three essential amino acids.
  • the liquid composition may comprise at least four essential amino acids.
  • the liquid composition may comprise at least five essential amino acids.
  • the liquid composition may comprise at least six essential amino acids.
  • the liquid composition may comprise at least seven essential amino acids.
  • the liquid composition may comprise at least eight essential amino acids.
  • the liquid composition may comprise at least nine essential amino acids.
  • the liquid composition may comprise at least ten essential amino acids.
  • the liquid composition comprises at least one vitamin.
  • the vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline.
  • the amount of vitamin is at least, approximately, or no more than about 0.00001, 0.0001, 0.001, 0.01, 0.1, 1, 25, 50, 75, 100, 125, 150, 175 or 200 ⁇ M.
  • the amount of vitamin may be approximately about 0.000010 to 200 ⁇ M.
  • the liquid composition comprises approximately at least 0.001, 100, or 1000 ⁇ M of at least one vitamin.
  • the vitamin is vitamin C.
  • the liquid composition comprises at least approximately 1 ⁇ M of the vitamin C.
  • the liquid composition may comprise at least approximately 7 ⁇ M of the vitamin C.
  • the liquid composition may comprise at least approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 ⁇ M of vitamin C.
  • the vitamin is vitamin B.
  • the liquid composition comprises 0.000005 to 1500 ⁇ M of said vitamin B, such as 0.00005, 0.0005, 0.005, 0.05, 0.5, 1, 10, 100, 1000 or 1500 ⁇ M.
  • Vitamin B may include biotin, vitamin B1, vitamin B2, vitamin B3, vitamin B5, vitamin B6, vitamin B9, and/or vitamin B12.
  • the liquid composition comprises glutamate.
  • the amount of glutamate may be at least, approximately, or no more than 1, 5, 20, 50, 100, 200, 300, 400, 500, 750, or 1000 ⁇ M.
  • the amount of glutamate may be approximately 2-500, 20-400, 75-300, or 100-200 ⁇ M.
  • the amount of glutamate increases glutamate in the blood by at least, approximately or no more than 25, 50, 75, 100, 125, 150, 175, 200, or 300 ⁇ M.
  • the amount of glutamate increases glutamate in the blood by at least 150 ⁇ M.
  • the liquid composition further comprises an essential fatty acid, such as omega-6 and omega-3 fatty acids.
  • the essential fatty acid is an omega-3 fatty acid.
  • the amount of essential fatty acid may be approximately 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1500, or 2000 mg per liter. In some embodiments, the amount of essential amino acids is 200-350 mg per liter.
  • the liquid composition further comprises at least one mineral.
  • the mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc.
  • the liquid composition comprises at least 5 nM of the mineral.
  • the amount of at least one of the mineral may be 5 nM-5 ⁇ M.
  • the liquid composition comprises 1 ⁇ M or more.
  • the liquid composition described herein comprises at least one ion.
  • the amount of at least one ion is at least approximately 0.1 mEq/L.
  • the amount of at least one ion may be 0.1-150 mEq/L.
  • the amount of at least one ion may be at least approximately 0.1, 0.5, 1, 5, 10, 15, 20, 50, 75, 100, 125, or 150 mEq/L.
  • the liquid composition is substantially free of at least one nonessential amino acid selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine.
  • the liquid composition may be substantially free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids.
  • the liquid composition may be free of at least one nonessential amino acid.
  • the liquid composition may be free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids.
  • the liquid composition may be free of nonessential amino acids other than glutamate.
  • the liquid composition further comprises at least one nonessential amino acid.
  • the nonessential amino acid can be selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, and tyrosine.
  • the liquid composition comprises at least two nonessential amino acids.
  • the liquid composition may comprise at least three nonessential amino acids.
  • the liquid composition may comprise at least four nonessential amino acids.
  • the liquid composition may comprise at least five nonessential amino acids.
  • the liquid composition may comprise at least six nonessential amino acids.
  • the liquid composition may comprise at least seven nonessential amino acids.
  • the liquid composition may comprise at least eight nonessential amino acids.
  • the liquid composition may comprise at least nine nonessential amino acids.
  • the amount of the nonessential amino acid is at least 5 approximately ⁇ M, such as 5, 10, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or 700 ⁇ M.
  • the amount of the nonessential amino acid may be 5-600 ⁇ M.
  • the liquid composition may comprise less than approximately 5 ⁇ M of cysteine and/or cystine.
  • the amount of cysteine and/or cystine may be 0, 0.1, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, or 5 ⁇ M.
  • the liquid composition comprises 0-4 ⁇ M of cysteine and/or cystine.
  • the liquid composition may comprise 0-2.5 ⁇ M of cysteine and/or cystine, such as 0-1 ⁇ M of cysteine and/or cystine.
  • the liquid composition is substantially free of cysteine and/or cystine.
  • the liquid composition may be free of cysteine and/or cystine.
  • the liquid composition has a pH of 5.0-6.5.
  • the liquid composition may have a pH of 5.5-6.0.
  • the liquid composition may have a pH of approximately 6.0.
  • the liquid composition may have a pH of approximately 5.5.
  • the liquid composition may have a pH of approximately 5.0.
  • the liquid composition has an osmolarity of 300-500.
  • the liquid composition may have an osmolarity of 300-400.
  • the liquid composition has an osmolarity of 300-350.
  • the liquid composition may have an osmolarity of 400-500.
  • the liquid composition contains an aqueous solution that may be a sterile solution or a nonsterile solution. In some embodiments, the aqueous solution is sterile. In other embodiments, the aqueous solution is nonsterile.
  • the liquid composition may be stored in a container that is ready to be used by the subject.
  • the container may comprise a sufficient amount of liquid composition for use in any of the methods described herein. For example, the container may hold approximately 1, 2, or 2.5 liter of liquid composition.
  • the liquid composition may be available as a ready-for-use dialysis solution.
  • the present disclosure provides dialysis compositions.
  • the dialysis composition may be in the form of a dry or concentrated composition.
  • the present disclosure provides a dry or concentrated composition comprising glutamate.
  • the dry or concentrated composition comprises glutamate and at least one essential amino acid.
  • the present disclosure also provides a dialysis composition comprising at least one vitamin.
  • the dry or concentrated composition comprises at least one vitamin and at least one essential amino acid.
  • the dry or concentrated composition is substantially free of cysteine and/or cystine.
  • the present disclosure provides, a dry composition
  • a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the dry composition optionally comprises glutamate, wherein the dry composition comprises 0-5 ⁇ M of cysteine and/or cystine.
  • the present disclosure provides, a dry composition
  • a dry composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one vitamin; (d) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (e) at least one essential amino acid, wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • the present disclosure provides a dry composition
  • a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the dry composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, and wherein the dry composition comprises 0-5 ⁇ M of cysteine and/or cystine.
  • the present disclosure provides, a dry composition
  • a dry composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (d) at least one essential amino acid, wherein the dry composition is substantially free of nonessential amino acids other than glutamate, and wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • the present disclosure provides a dry composition
  • a dry composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (c) at least one essential amino acid, wherein the dry composition comprises at least 2 ⁇ g essential amino acid, and wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • the present disclosure provides a dry composition comprising: (a) carbohydrate; (b) at least one essential amino acid, wherein the dry composition comprises 0-10 ⁇ g of cysteine and/or cystine.
  • the dry composition described herein may be advantageous since it may minimize storage space, weight, and transport of water and may also increase the stability and thereby also the shelf-life of the dialysis components.
  • the dry composition may be stable for long term storage and transportation.
  • the dry composition may be available in a container, in which an aqueous solution may be added to the container to reconstitute the contents therein.
  • the container may have the capacity to hold approximately 1, 2, 2.5, 4 or 10 liter of aqueous solution.
  • Aqueous solution may be added to the container prior to the administration of the composition to the subject for dialysis.
  • the carbohydrate is selected from a group consisting of dextrose, glucose, sucrose, fructose, and lactose.
  • the carbohydrate is dextrose.
  • the amount of dextrose in the dry composition may range from 0.1 g to 75 g. In certain embodiments, the amount of dextrose is approximately 5-75 g. The amount of the dextrose may be approximately 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, or 75 g.
  • the amount of the dextrose may be 10-50 g. In one embodiment, the amount of dextrose is approximately 10 g. In one embodiment, the amount of dextrose is approximately 50 g.
  • the amount of dextrose is 0.1 g-5 g.
  • the amount of the dextrose may be approximately 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5 or 5.0 g.
  • the amount of the dextrose may be 0.5-2.5 g.
  • the amount of dextrose is approximately 0.5 g.
  • the amount of dextrose is approximately 2 g.
  • the amount of dextrose is approximately 10-99.5% of the total composition.
  • the amount of dextrose may be 10, 20, 30, 40, 50, 60, 70, 75, 80, 85, 90, 95, or 99.5% of the total composition.
  • the amount of dextrose is 70-95% of the total composition.
  • the dry composition comprises at least one essential amino acid selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine.
  • at least one of the essential amino acid is methionine.
  • at least one of the essential amino acid is arginine.
  • the amount of each essential amino acid is at least 2 ⁇ g.
  • the amount of each essential amino acid may be at least, approximately, or no more than 1, 2, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 300, 500, 1000, or 1500 ⁇ g.
  • the total amount of the essential amino acids may be at least, approximately, or no more than 100, 150, 200, 250, 300, 500, 1000, 2000, 3000, 5000, 6000, 7000, 7500 or 8000 ⁇ g.
  • the dry composition comprises at least two essential amino acids.
  • the dry composition may comprise at least three essential amino acids.
  • the dry composition may comprise at least four essential amino acids.
  • the dry composition may comprise at least five essential amino acids.
  • the dry composition may comprise at least six essential amino acids.
  • the dry composition may comprise at least seven essential amino acids.
  • the dry composition may comprise at least eight essential amino acids.
  • the dry composition may comprise at least nine essential amino acids.
  • the dry composition may comprise at least ten essential amino acids.
  • the dry composition comprises at least one vitamin.
  • the vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline.
  • the amount of vitamin is at least, approximately, or no more than 0.01, 0.05, 0.1, 0.5, 1, 10, 50, 100, 500, 750, 1000, 1250, or 1500 ⁇ g.
  • the amount of vitamin may be approximately 0.01-1500 ⁇ m.
  • the vitamin is vitamin C.
  • the dry composition may comprise at least approximately 1000 ⁇ g of vitamin C, such as approximately 1000, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, or 3000 ⁇ g of vitamin C.
  • the dry composition comprises at least approximately 2500 ⁇ g of vitamin C.
  • the dry composition may comprise at least, approximately, or no more than 0.01 ⁇ g of vitamin B, such as approximately 0.01, 0.05, 0.1, 0.5, 1, 1.5, 5, 10, 50, or 100 ⁇ g of vitamin B.
  • Vitamin B may include biotin, vitamin B1, vitamin B2, vitamin B3, vitamin B5, vitamin B6, vitamin B9, and/or vitamin B12.
  • the dry composition comprises at least, approximately, or no more than 0.1 mg of glutamate, such as approximately 0.1, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 mg.
  • the amount of glutamate may be 0.1-50 mg, 5-25 mg, or 25-45 mg.
  • the dry composition further comprises an essential fatty acid, such as omega-6 and omega-3 fatty acids.
  • the essential fatty acid is an omega-3 fatty acid.
  • the essential fatty acid is an omega-3 fatty acid.
  • the amount of essential fatty acid may be approximately 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 mg per liter. In some embodiments, the amount of essential amino acids is 1200-1600 mg per liter.
  • the dry composition further comprises at least one mineral.
  • the mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc.
  • the dry composition comprises at least 0.3 ⁇ g of the mineral.
  • the dry composition comprises approximately 0.1, 1, 10, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1250, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000 or 7500 ⁇ g.
  • the dry composition comprises approximately 400 ⁇ g.
  • the dry composition comprises approximately 800 ⁇ g.
  • the dry composition described herein comprises at least one ion.
  • the ion may be, but are not limited to, sodium, calcium, magnesium, chloride, lactate or combinations thereof.
  • the amount of at least one of the ion is at least approximately 0.1 ⁇ g. In one embodiment, the amount of at least one of the ion is at least approximately 0.5 ⁇ g.
  • the amount of the ion may be at least approximately 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 ⁇ g. In certain embodiments, the amount of the ion is at least approximately 50 ⁇ mol.
  • the amount of ion is at least approximately 100 ⁇ mol.
  • the amount of ion may be at least approximately 200 ⁇ mol.
  • the amount of ion may be at least approximately 250 ⁇ mol.
  • the amount of the ion may be 50-300 ⁇ mol, such as 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, or 300 ⁇ mol.
  • the dry composition is substantially free of at least one nonessential amino acid selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine.
  • the dry composition may be substantially free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids.
  • the dry composition may be free of at least one nonessential amino acid.
  • the dry composition may be free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids.
  • the dry composition may be free of nonessential amino acids other than glutamate.
  • the dry composition further comprises at least one nonessential amino acid.
  • the nonessential amino acid can be selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, and tyrosine.
  • the dry composition comprises at least two nonessential amino acids.
  • the dry composition may comprise at least three nonessential amino acids.
  • the dry composition may comprise at least four nonessential amino acids.
  • the dry composition may comprise at least five nonessential amino acids.
  • the dry composition may comprise at least six nonessential amino acids.
  • the dry composition may comprise at least seven nonessential amino acids.
  • the dry composition may comprise at least eight nonessential amino acids.
  • the dry composition may comprise at least nine nonessential amino acids.
  • the amount of the nonessential amino acid is at least 1 mg.
  • the amount of the nonessential amino acid may be 1-100 mg.
  • the amount of nonessential amino acid is 1-300 mg, such as 1, 50, 100, 150, 200, 250, or 300 mg.
  • the present disclosure provides a dry composition comprising less than 10 mg of cysteine and/or cystine.
  • the dry composition may comprise approximately 0, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 or 10 mg of cysteine and/or cystine.
  • the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • the dry composition comprises 0-2.5 mg of cysteine and/or cystine.
  • the dry composition is substantially free of cysteine and/or cystine.
  • the dry composition may be free of cysteine and/or cystine.
  • the liquid composition or dry composition comprises methionine.
  • the molar ratio of dextrose to methionine of the liquid composition or dry composition may be greater or less than approximately 1, 50, 100, 250, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 10,500, 20,000, 30,000, 40,000, 50,500, 60,000, 70,000, 80,000, 90,000, or 100,000.
  • the molar ratio of dextrose to methionine of the liquid composition or dry composition is greater or less than approximately 250, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, or 10,500.
  • the molar ratio of dextrose to methionine of the compositions is approximately 250.
  • the molar ratio of dextrose to methionine of the compositions is approximately 10,500.
  • the molar ratio of cysteine to essential amino acids of the liquid composition or dry composition may be less than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 10, 15, 20, 50, or 100.
  • the molar ratio of cysteine to methionine is less than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to amino acids of the liquid composition or dry composition may be greater than about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 10, 15, 20, 50, or 100.
  • the molar ratio of methionine to other amino acids of the liquid composition and dry composition may be greater than about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to alanine in the liquid composition or dry composition is greater than about 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to arginine is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to asparagine is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to aspartic acid is greater than about 2, 3, 4, 5, 10, 15, 20, 50, or 100. In some embodiments, the molar ratio of methionine to ⁇ -alanine is greater than about 1, 2, 3, 4, 5, 10, 15, 20, 50, or 100. In some embodiments, the molar ratio of methionine to cystine is greater than about 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to glutamic acid is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to glutamine is greater than about 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to glycine is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to histidine is greater than about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to isoleucine is greater than about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to leucine is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to phenylalanine is greater than about 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to proline is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to serine is greater than about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to threonine is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to tryptophan is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to tyrosine is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to valine is greater than about 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • the molar ratio of methionine to minerals in the dry composition or liquid composition, such as iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium and zinc, of the liquid composition or dry composition is greater than about 1, 5, 7, 8, 9, 10, 40, 50, 75, 90, 100, 500, 1,000, 1,100, 1,500, 1,900, 2,000, 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000.
  • the molar ratio of methionine to iron may be greater than about 9, 10, 40, 50, 75, 90, 100, 500, or 1,000.
  • the molar ratio of methionine to copper may be greater than about 90, 100, 500, 1,000, 1,100, 1,500, 1,900, or 2,000.
  • the molar ratio of methionine to chromium may be greater than about 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000.
  • the molar ratio of methionine to fluoride may be greater than about 7, 8, 9, 10, 40, 50, 75, 90, 100, 500, or 1,000.
  • the molar ratio of methionine to iodide may be greater than about 1,100, 1,500, 1,900, 2,000, 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000.
  • the molar ratio of methionine to manganese may be greater than about 40, 50, 75, 90, 100, 500, or 1,000.
  • the molar ratio of methionine to molybdenum may be greater than about 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000.
  • the molar ratio of methionine to selenium may be greater than about 1,900, 2,000, 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000.
  • the molar ratio of methionine to zinc may be greater than about 8, 9, 10, 40, 50, 75, 90, 100, 500, or 1,000.
  • the dry composition, concentrated composition or liquid composition comprises one or more components, such as dextrose, ions, buffer, amino acids, vitamins and minerals, in terms of molar ratio (dextrose to component), as depicted in Table 1 below.
  • the dry composition may also be available in a concentrated form.
  • the present disclosure provides a concentrated composition comprising a dry composition described herein, and an initial aqueous solution.
  • the concentrated composition described herein may be advantageous since it may minimize storage space, weight, and transport of water and may also increase the stability and thereby also the shelf-life of the dialysis components.
  • the concentrated composition may be stable for long term storage and transportation.
  • the concentrated composition may be available in a container, in which additional aqueous solution may be added to the container to bring the solution to volume, at which point, the concentrated composition may be ready to use by the subject.
  • the container may have the capacity to hold approximately 1, 2, 2.5, 4, or 10 liter of total aqueous solution.
  • the additional aqueous solution may be added to the concentrated composition prior to administration to a subject for dialysis.
  • the present disclosure provides a food composition
  • a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least one nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline, wherein each nonessential amino acid is 0-1% wt/wt of protein of the food composition, wherein the food composition comprises 0-1% wt/wt of cysteine and/or cystine of protein of the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • the amount of each nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline, is 0-2% wt/wt of the protein in the food composition.
  • the amount of each nonessential amino acid may be approximately 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0% wt/wt of the protein.
  • the amount of each nonessential amino acid may be 0-1% wt/wt of the protein in the food composition. In one embodiment, the amount of each nonessential amino acid is 0-0.1% wt/wt of the protein in the food composition. In some embodiments, the food composition comprises at least two of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least three of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least four of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least five of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • the food composition may comprise at least six of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline.
  • at least one of the nonessential amino acid is glycine.
  • at least one of the nonessential amino acid is proline.
  • the amount of the cysteine and/or cystine is 0-2% wt/wt of the protein in the food composition.
  • the amount of the cysteine and/or cystine may be 0-1% wt/wt of the protein in the food composition, such as 0, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1% wt/wt of the protein.
  • the amount of cysteine and/or cystine is 0-0.2% wt/wt of the protein in the food composition.
  • the amount of cysteine and/or cystine is approximately 0-0.1% wt/wt of the protein in the food composition. In another embodiment, the amount of cysteine and/or cysteine is 0-0.04% wt/wt of the food composition, such as approximately 0, 0.01, 0.02, 0.03, or 0.04% wt/wt of the food composition.
  • the present disclosure provides a food composition
  • a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least six nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine, wherein each nonessential amino acid is 0-1% wt/wt of protein in the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • the present disclosure provides a food composition comprising: (a) at least one essential amino acid; and (b) glutamate, wherein the food composition is substantially free of nonessential amino acids other than glutamate.
  • the present disclosure provides a food composition comprising: (a) at least one essential amino acid; (b) glutamate; and (c) arginine, wherein the food composition is substantially free of nonessential amino acids other than glutamate.
  • the amount of each nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine is 0-2% wt/wt of the protein in the food composition.
  • the amount of each nonessential amino acid may be approximately 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0% wt/wt of the protein.
  • the amount of each nonessential amino acid may be 0-1% wt/wt of the protein in the food composition. In one embodiment, the amount of each nonessential amino acid is 0-0.1% wt/wt of the protein in the food composition.
  • the food composition comprises at least seven of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least eight of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least nine of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least ten of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • the food composition may comprise at least six of the nonessential amino acids are selected from the group consisting of glycine, proline, glutamine, serine, tyrosine, and cysteine.
  • At least one of the nonessential amino acid is glycine. In some embodiments, at least one of the nonessential amino acid is proline. In an exemplary embodiment, at least one of the nonessential acids is cysteine.
  • the amount of cysteine and/or cystine is 0-2% wt/wt of protein in the food composition. The amount of the cysteine and/or cystine may be approximately 0-1% wt/wt of the protein in the food composition, such as approximately 0, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1% wt/wt of the protein.
  • the amount of cysteine and/or cystine is 0-0.2% wt/wt of the protein in the food composition. In one embodiment, the amount of cysteine and/or cystine is 0-0.1% wt/wt of the protein in the food composition. In another embodiment, the amount of cysteine and/or cysteine is 0-0.04% wt/wt of the food composition, such as approximately 0, 0.01, 0.02, 0.03, or 0.04% wt/wt of the food composition. In an exemplary embodiment, the food composition is substantially free of cysteine and/or cystine. The food composition may be free of cysteine and/or cystine.
  • the amount of the cysteine devoid protein source is at least 1% wt/wt of the food composition.
  • the cysteine devoid protein source may be gelatin.
  • the amount of the a cysteine devoid protein source may be 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, or 10.0% wt/wt of the food composition.
  • the amount of the cysteine devoid protein source is 1-5% of the food composition.
  • the amount of protein is at least approximately 5% of the total weight of the food composition.
  • the amount of protein in the food composition may be approximately 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25% of the total weight of the food composition.
  • the amount of protein is approximately 6% of the total weight of the food composition. In another embodiment, the amount of protein is approximately 16% of the total weight of the food composition.
  • the essential amino acid is selected from a group consisting of arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.
  • the essential amino acid is methionine.
  • the essential amino acid is arginine.
  • the essential amino acid may be approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, or 75% wt/wt of the food composition.
  • the essential amino acid is at least approximately 2% wt/wt of the food composition.
  • the amount of the essential amino acid may be 2-30% wt/wt of the food composition. In an exemplary embodiment, the amount of the essential amino acid is approximately 8% wt/wt of the food composition.
  • the food composition comprises at least two essential amino acids.
  • the food composition may comprise at least three essential amino acids.
  • the food composition may comprise at least four essential amino acids.
  • the food composition may comprise at least five essential amino acids.
  • the food composition may comprise at least six essential amino acids.
  • the food composition may comprise at least seven essential amino acids.
  • the food composition may comprise at least eight essential amino acids.
  • the food composition may comprise at least nine essential amino acids.
  • the food composition may comprise at least ten essential amino acids.
  • any food composition described herein may further comprise at least one vitamin.
  • the vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline.
  • the vitamin is vitamin C.
  • the food composition comprises at least approximately 10 mg of vitamin C per kg of the food composition.
  • the food composition may comprise approximately 10, 15, 20, 25, 30, 40, 45, 50, 55, or 60 mg of vitamin C per kg of the food composition.
  • the food composition comprises approximately 40 mg of vitamin C per kg of the food composition.
  • any food compositions described herein further comprises an essential fatty acid, such as omega-6 and omega-3 fatty acids.
  • the essential fatty acid is an omega-3 fatty acid.
  • the amount of essential fatty acid may be approximately 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 mg per liter.
  • the amount of essential amino acids is 1200-1600 mg per liter.
  • the amount of essential amino acids is 100-200 mg per liter.
  • any food compositions described herein further comprises at least one mineral.
  • the mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc.
  • the food composition comprises at least approximately 1% w/w of mineral, such as 1, 2, 3, 4, or 5% w/w of mineral in the food composition.
  • the amount of at least one of the mineral may be 0.05 mg-20 g per kg of the food composition, such as approximately 0.1, 1, 10, 50, 100, 500, 1,000, 5,000, 10,000 or 20,000 mg of mineral per kg of the food composition.
  • any food compositions described herein further comprise at least one ion.
  • the amount of at least one ion is at least 0.1% w/w, such as 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0% w/w of ion in the food composition.
  • the amount of at least one ion may be 0.05-20 g per kg food, such as approximately 0.05, 0.1, 1, 5, 10, 15, or 20 g of ion per kg of the food composition.
  • any food compositions described herein further comprise glutamate.
  • the food composition comprises at least, approximately, or no more than 0.1 mg of glutamate, such as approximately 0.1, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 mg.
  • the amount of glutamate may be 0.1-50 mg, 5-25 mg, or 25-45 mg.
  • any of the food compositions described herein comprise at least approximately 10% wt/wt of amino acids, such as 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20% w/w or amino acids.
  • the food composition comprises approximately 14% wt/wt of amino acids.
  • the food composition comprises approximately 17% wt/wt of amino acids.
  • the food composition comprises 10-20% w/w of total amino acids.
  • the food composition may comprise 14-17% w/w of total amino acids.
  • the food composition further comprises a pharmaceutically active agent.
  • the amount of pharmaceutical agent, or any other component used in the food compositions described herein can be used in an amount that is therapeutically effective.
  • the therapeutic amount can be assessed for individual subjects, or for groups of subjects.
  • the group of subjects can be all potential subjects, or subjects having a particular characteristic such as age, weight, race, gender, or physical activity level.
  • the pharmaceutically active agent may include anti-cancer therapeutics, such as mesna.
  • the pharmaceutically active agent provided to the subject may be approximately 5, 10, 50, 100, 500, 1000, 1200, 1500, 2000, 2500, or 3000 mg.
  • the pharmaceutically active agent provided to the subject is 10-2000 mg.
  • the mesna is provided to the subject is 200-1200 mg.
  • the pharmaceutically active agent is an anti-obesity therapeutic.
  • the invention provides for methods of treating (including preventing) cancer, metabolic diseases, such as obesity, and/or renal disease in subjects in need thereof.
  • the method can include the reduction, depletion and/or supplementation of certain nutrients to the subject's diet.
  • the method can include administering to the subject a food composition described herein that has no more than certain amounts of pre-selected nutrients.
  • the nutrients that are reduced or depleted can be certain nutrients that are demanded at a higher level or degree by cancerous cells as compared to non-cancerous cells.
  • the subject's diet can be controlled by administering to the subject a food composition wherein the subject does not eat any other foods or nutrients other than those that are part of the food composition, or permitted under the regimen.
  • the food composition may consist of all of the foods provided to the subject, and the food composition may have only certain amounts of pre-selected nutrients.
  • the food composition can be designed such that the nutrients provided to or ingested by the subject are evaluated or determined on an hourly, daily, weekly or monthly basis. For any numbers provided herein that are described on a daily basis, the invention provides for proportionally scaled numbers for such amounts on an hourly, weekly or monthly basis.
  • the food composition can be provided to the subject, alone or in combination with the dry composition, concentrated composition or liquid composition, until consistent and significant improvement is seen in the subject, and may be modified at any point of the treatment to fit the needs of the subject.
  • the methods described herein also include the reduction, depletion and/or supplementation of certain nutrients in the subject's system, such as blood levels, through dialysis.
  • the present disclosure envisions methods of performing dialysis.
  • Dialysis machines for performing dialysis such as Freedom Cycler (Fresenius Medical Care), HomeChoice PROTM (Baxter Healthcare Corporation), HomeChoiceTM (Baxter Healthcare Corporation), Liberty® Cycler (Fresenius Medical Care), NewtonTM IQ Cycler System (Fresenius Medical Care), 2008K@HomeTM (Fresenius Medical Care), NxStage® System OneTM with PureFlowTM SL (NxStage Medical, Inc.), Quanta SC+(Quanta Dialysis Technologies) and the like are known in the art, and may be used to carry out the methods described herein.
  • the methods can include administering to the subject a liquid composition, concentrated composition, or dry composition described herein that has no more than certain pre-selected nutrients, which can be used alone or in combination with a diet that administers a food composition.
  • the methods comprise placing a subject in need thereof on a nutritional diet with any of the food compositions disclosed herein and on dialysis with a dialysate composition (liquid composition, dry composition, or concentrated composition).
  • the subject is placed on a nutritional diet before, during, and/or after dialysis treatment.
  • the nutrients that are reduced or depleted can be certain nutrients that are demanded at a higher level or degree by cancerous cells as compared to non-cancerous cells.
  • the levels of these nutrients can be controlled by administering to the subject a dry composition, concentrated composition, or liquid composition and periodically monitoring the subject.
  • the methods may involve administering the composition to draw waste, fluid, and/or certain nutrients, such as cysteine and/or cystine, from the subject's blood.
  • the dry composition, concentrated composition, or liquid composition may consist of a substantial amount of nutrients necessary for the subject, such as one or more essential amino acids, arginine, and glutamate.
  • the methods may also involve administering the composition to provide the subject with necessary nutrients.
  • the dialysate composition may be in the subject body as long necessary for the composition to exchange waste, fluids, and/or nutrients with the body.
  • the exchange (drain and refill) may be facilitated by the subject or with the assistance of a machine. In some embodiments, the exchange takes at least 15, 20, 25, 30, 35, 40, 45, 50, 55, or 60 minutes.
  • the dry composition, concentrated composition or liquid composition can be designed such that the nutrients provided to the subject are evaluated or determined on an hourly, daily, weekly or monthly basis. For any numbers provided herein that are described on a daily basis, the invention provides for proportionally scaled numbers for such amounts on an hourly, weekly or monthly basis.
  • the dry composition, concentrated composition, or liquid composition, alone or in combination with a food composition can be provided to the subject until consistent and significant improvement is seen in the subject, and may be modified at any point of the treatment to fit the needs of the subject.
  • Peritoneal dialysis may involve the insertion of a flexible tube, such as a catheter, into the subject.
  • the catheter may go inside the subject's abdomen or chest to outside the subject's body through an incision in the subject's skin.
  • the catheter can fill the abdomen with dialysate
  • FIG. 14 illustrates an exemplary dialysis system.
  • the subject undergoes hemodialysis.
  • the methods described herein may comprise providing a composition described herein.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • treating cancer is characterized by a reduction in tumor size.
  • the tumor size may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial tumor size prior to treatment.
  • the tumor may be reduced by at least 30%.
  • the tumor may be reduced by at least 50%.
  • the tumor may be reduced by at least 80%.
  • the reduction of tumor size may be 10-100%.
  • the reduction of tumor size may be 30-100%.
  • treating cancer is characterized by a reduction in circulated cancer cell counts.
  • the circulated cancer cell count may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial circulated cancer cell count prior to treatment.
  • the circulated cancer cell count may be reduced by at least 30%.
  • the circulated cancer cell count may be reduced by at least 50%.
  • the circulated cancer cell count may be reduced by at least 80%.
  • the reduction of circulated cancer cell count may be 10-100%.
  • the reduction of circulated cancer cell count may be 30-100%.
  • the methods of the present disclosure include monitoring levels of cysteine and/or cystine in the subject's blood. Monitoring cysteine and/or cystine may be used to modify the existing treatment in order to reduce the levels of cysteine and/or cystine to therapeutic levels.
  • treating cancer is characterized by decreased levels of plasma cysteine.
  • the levels of plasma cysteine may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial levels of plasma cysteine prior to treatment.
  • the levels of plasma cysteine may be reduced by at least 30%.
  • the levels of plasma cysteine may be reduced by at least 50%.
  • the levels of plasma cysteine may be reduced by at least 80%.
  • the levels of plasma cysteine may be reduced by at least 90%.
  • the levels of plasma cysteine may be reduced by at least 95%.
  • the levels of plasma cysteine may be 30-100%.
  • Administering a composition described herein may result in a reduction of blood levels of cysteine and/or cystine.
  • the composition may be administered in therapeutic amounts.
  • the level of cysteine in the subject's blood may be reduced to less than average levels of cysteine, such as less than approximately 10 ⁇ M of cysteine.
  • the level of cysteine in the subject's blood is less than approximately 9 ⁇ M, 8 ⁇ M, 7 ⁇ M, 6 ⁇ M, 5 ⁇ M, 4 ⁇ M, 3 ⁇ M, 2 ⁇ M, or 1 ⁇ M
  • the level of cysteine in the subject's blood is approximately less than 1 ⁇ M.
  • the level of cystine in the subject's blood may be reduced to less than average levels of cystine, such as less than approximately 40 ⁇ M of cystine.
  • the level of cystine in the subject's blood may be reduced to less than approximately 39 ⁇ M, 38 ⁇ M, 37 ⁇ M, 36 ⁇ M, 35 ⁇ M, 34 ⁇ M, 33 ⁇ M, 32 ⁇ M, 31 ⁇ M, 30 ⁇ M, 29 ⁇ M, 28 ⁇ M, 27 ⁇ M, 26 ⁇ M, 25 ⁇ M, 24 ⁇ M, 23 ⁇ M, 22 ⁇ M, 21 ⁇ M, 20 ⁇ M, 19 ⁇ M, 18 ⁇ M, 17 ⁇ M, 16 ⁇ M, 15 ⁇ M, 14 ⁇ M, 13 ⁇ M, 12 ⁇ M, 11 ⁇ M, 10 ⁇ M, 9 ⁇ M, 8 ⁇ M, 7 ⁇ M, 6 ⁇ M, 5 ⁇ M, 4 ⁇ M, 3
  • the present disclosure provides a method for reducing tumor size and/or circulated cancer cell counts in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • the present disclosure provides a method for reducing tumor size and/or circulated cancer cell counts in a subject in need thereof, the method comprising administering to the subject a liquid composition of claim 1 , a dry composition described herein, or a concentrated composition described herein.
  • the present disclosure provides a method for reducing tumor size and/or circulated cancer cell counts in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • Methods of the disclosure may result in suppressing tumor growth in a subject.
  • the subject's tumor size does not increase by greater than 0-100%, such as approximately 0, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100%, as measured before and 6 weeks after the initial administration of any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • the subject's tumor size may not increase by greater than approximately 100%, as measured before and 6 weeks after the initial administration of any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • the subject's tumor size may not increase by greater than approximately 70%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may not increase by greater than approximately 60%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may not increase by greater than approximately 40%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may not increase by greater than approximately 20%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may not increase by greater than approximately 0%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size decreases by at least 10%, as measured before and 6 weeks after the initial administration of any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • the subject's tumor size may decrease by at least 20%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may decrease by at least 30%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may decrease by at least 40%, as measured before and 6 weeks after the initial administration.
  • the subject's tumor size may decrease by at least 50%, as measured before and 6 weeks after the initial administration.
  • Methods of the disclosure may result in a reduction in circulated cancer cell counts.
  • the circulated cancer cell count may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial circulated cancer cell count prior to treatment.
  • the circulated cancer cell count may be reduced by at least 30%.
  • the circulated cancer cell count may be reduced by at least 50%.
  • the circulated cancer cell count may be reduced by at least 80%.
  • the reduction of circulated cancer cell count may be 10-100%.
  • the reduction of circulated cancer cell count may be 30-100%.
  • the present disclosure provides a method for treating obesity in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • Treating obesity may characterized by weight loss in a subject.
  • the subject may experience of at least or approximately 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50% weight loss from the start of the nutritional diet with the food compositions disclosed herein.
  • the subject's weight loss is greater than 5%, as measured before and 6 weeks after commencement of the controlled diet.
  • a controlled diet comprises administering a food composition described herein to a subject in need thereof.
  • the subject's weight loss may be greater than 10%, as measured before and 6 weeks after commencement of the controlled diet.
  • the subject's weight loss may be greater than 15%, as measured before and 6 weeks after commencement of the controlled diet.
  • the subject's weight loss may be greater than 20%, as measured before and 6 weeks after commencement of the controlled diet.
  • the subject's weight loss may be greater than 25%, as measured before and 6 weeks after commencement of the controlled diet.
  • the subject's weight loss may be greater than 30%, as measured before and 6 weeks after commencement of the controlled diet.
  • the present disclosure provides a method for treating renal disease in a subject in need thereof, the method comprising administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition.
  • the methods of the present disclosure may provide or deplete certain nutrients to the subject.
  • the subject may be experiencing malnutrition, nutrition deficiency or other side effects from dialysis, and administration of a liquid composition, dry composition, or concentrated composition may prevent, alleviate, reduce, or eliminate such side effects.
  • Other side effects may include, but are not limited to, low blood pressure, nausea, vomiting, dry and itchy skin, restless leg syndrome, muscle cramping, or combinations thereof.
  • the methods described herein administering a liquid composition or dry composition in certain embodiments, the methods further comprise utilizing a membrane.
  • the membrane may comprise modified cellulose diacetate or polysulfone.
  • the membrane is a low-flux membrane that permits small molecule exchange.
  • the low-flux membrane comprises a plurality of pores, in which the plurality of pores comprises a first end and a second end.
  • the low-flux membrane comprises a plurality of pores.
  • the plurality of pores may have an average pore diameter of approximately 1.8 nm.
  • the low-flux membrane permits approximately 50% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end.
  • the low-flux membrane may permit approximately 75% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. In some embodiments, the low-flux membrane permits approximately 90% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. The low-flux membrane may permit approximately 95% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. In some embodiments, approximately 20% of a molecule of at least 1000 Daltons passes through the plurality of pores, from the first end to the second end. Approximately 10% of a molecule of at least 1000 Daltons may pass from the first side to the second side. Approximately 1% of a molecule of at least 1000 Daltons may pass from the first side to the second side.
  • the method can further comprise administering a pharmaceutically active agent.
  • the pharmaceutically active agent may be an anti-cancer therapeutic.
  • anti-cancer drugs include Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino-17-demethoxygeldanamycin, Alpharadin, Alvocidib, 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone, Amonafide, Anthracenedione, Anti-CD22 immunotoxins, Antineoplastic, Antitumorigenic herbs, Apaziquone, Atiprimod, Azathioprine, Belotecan, Bendamustine, BMW 2992, Biricodar, Brostallicin, Bryostatin, Buthionine sulfoxi
  • the pharmaceutically active agent may be an anti-obesity therapeutic.
  • anti-obesity therapeutic agents include lipase inhibitors (such as Orlistat), dopaminergic, noradrenergic, and serotoninergic compounds, cannabinoid receptor antagonists (such as rimonabant), exenatide, pramlintide, and CNS agents (such as topimerate).
  • the pharmaceutically active agent is provided in a subtherapeutic amount.
  • the amount of pharmaceutical agent, or any other component used in a combination composition described herein, can be used in an amount that is subtherapeutic.
  • using sub-therapeutic amounts of an agent or component can reduce the side effects of the agent.
  • Use of subtherapeutic amounts can still be effective, particularly when used in synergy with other agents or components.
  • a subtherapeutic amount of the agent or component can be such that it is an amount below which would be considered therapeutic.
  • FDA guidelines can suggest a specified level of dosing to treat a particular condition, and a subtherapeutic amount would be any level that is below the FDA suggested dosing level.
  • the subtherapeutic amount can be about 1, 5, 10, 15, 20, 25, 30, 35, 50, 75, 90, or 95% less than the amount that is considered to be a therapeutic amount.
  • the therapeutic amount can be assessed for individual subjects, or for groups of subjects.
  • the group of subjects can be all potential subjects, or subjects having a particular characteristic such as age, weight, race, gender, or physical activity level.
  • the methods can further comprise administering a nutritional supplement.
  • the nutritional supplement comprises one or more nutrients, which may include vitamins, minerals, water, carbohydrates, fats, proteins, and combinations thereof.
  • Nutritional supplements may be packaged as vitamins, minerals, herbs, meal supplements, sports nutrition products, natural food supplements, and fortified food products.
  • the nutritional supplement may comprise at least one amino acid selected from a group consisting of arginine, histidine, isoleucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.
  • the nutritional supplement may provide the subject with nutrients at levels in accordance with the Dietary Reference Intake (DRI), the Reference Daily Intake (RDI), the World Health Organization (WHO) guidelines, Recommended Dietary Allowances (RDAs), Adequate Intakes (Ais), and/or Daily Value (DV).
  • the required levels of nutrients may be the same or different for adults, children, men, and women.
  • the nutritional supplement comprises one or more nutrients selected from a group consisting of vitamin A, thiamine, riboflavin, niacin, pantothenic acid, vitamin B6, biotin, folate, cyanocobalamin, vitamin C, vitamin D, vitamin E, vitamin K, and choline.
  • the nutritional supplement comprises one or more nutrients selected from a group consisting of calcium, chloride, chromium, copper, fluoride, iodine, iron, magnesium, manganese, molybdenum, phosphorus, potassium, selenium, sodium and zinc.
  • the nutritional supplement comprises one or more nutrients selected from a group consisting of water, carbohydrates, protein, fiber, fat, cholesterol, and fatty acids.
  • the present disclosure provides methods of providing dialysis therapies to subjects in need thereof.
  • the liquid composition, dry composition, or concentrated composition may be administered via traditional dialysis methods.
  • the compositions may be administered by hemodialysis, peritoneal dialysis, hemofiltration, hemodiafiltration, and intestinal dialysis.
  • the compositions are administered by hemodialysis or peritoneal dialysis (such as continuous ambulatory PD (CAPD) and continuous cycler-assisted PD (CCPD).
  • the dialysis treatment may be used in combination with other conventional methods for treating cancer, obesity and renal disease, such as surgery, radiation therapy, immunotherapy, anti-cancer therapeutics, anti-obesity therapeutics, or renal disease therapeutics.
  • the composition is administered to the subject at least once a week.
  • the composition may be administered to the subject at least three times a week.
  • the composition may be administered to the subject at least five times a week.
  • the composition is administered to the subject over the course of at least one week.
  • the composition may be administered to the subject over the course of at least four weeks.
  • the composition may be administered to the subject over the course of at least 2 months.
  • the composition may be administered to the subject over the course of at least 3 months.
  • the dialysis may be provided to a subject in need thereof in a dialysis center, a hospital, a clinic, health care facility, at home, or any place that is clean and dry.
  • any food composition described herein may be administered multiple times a week, such as 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more times a week.
  • the food composition is administered to the subject at least once a week.
  • the food composition may be administered to the subject at least three times a week.
  • the food composition may be administered to the subject at least five times a week.
  • the food composition may be administered to the subject at least twenty-one times a week.
  • the food composition may be administered to the subject over the course of at least one week.
  • the food composition may be administered to the subject over the course of at least four weeks.
  • the food composition may be administered to the subject over the course of at least 2 months.
  • the food composition may be administered to the subject over the course of at least 3 months.
  • the liquid composition, the dry composition or concentrated composition described herein is administered to the subject sequentially with any food composition described herein.
  • the liquid composition, the dry composition or concentrated composition described herein is administered to the subject simultaneously with any food composition described herein.
  • the food composition, and the liquid composition, dry composition or concentrated composition are administered to the subject at least once a week.
  • the food composition, and the liquid composition, dry composition or concentrated composition may be administered to the subject at least three times a week.
  • the food composition, and the liquid composition, dry composition or concentrated composition may be administered to the subject over the course of at least one week.
  • the food composition, and the liquid composition, dry composition or concentrated composition may be administered to the subject over the course of at least four weeks.
  • any food composition described herein may have a pre-selected caloric value.
  • the food composition may comprise at least 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 1500 kcal.
  • the food composition may have a caloric value 0-100, 0-200, 200-400, 400-600, 600-800, 800-1000 or 500-1000 kcal.
  • the food composition may have a mass that is at least 5, 10, 50, 100, 200, 300, 400, 500, 600, 700, or 1000 grams.
  • the food composition may have a mass that is 0-100, 100-200, 200-400, 400-600, or 100-500 grams.
  • any food composition as described herein may be sold to a subject in a packaged container.
  • the food composition is packaged as a solid food, semi-solid food, or beverage.
  • the food composition may be a concoction of nutrient ingredients, an instant quick meal mix, a microwaveable frozen paste, a bake- or roast-ready dough, or a customizable mixture of several nutrient modules with flavor varieties.
  • the food compositions may be cooked, partially cooked, or uncooked.
  • the food composition may be baby formula, a soft drink, pulp-free juice, a nutritional shake, a granola bar, a dessert, or candy.
  • the food composition is fortified with vitamins, minerals, herbs, nutrients, and combinations thereof.
  • the food composition may be packaged as a food product selected from a group consisting of a snack bar, cereal product, bakery product, and dairy product.
  • the food composition may be baby formula or baby food.
  • the food composition may be a soft drink.
  • the food composition may be a nutritional shake or nutritional snack, such as a granola bar.
  • the food composition may be meal replacement drink or snack.
  • the food composition may have a palatability enhancer.
  • the palatability enhancer may be can be MSG, or some other form of glutamate, salt.
  • the food composition is a pet food.
  • the pet food can be a wet or dry pet food.
  • the pet food can be formed as a kibble which may be coated.
  • the coating may be a polymer coating that is applied to the kibble after the kibble is formed.
  • additives, fillers or excipients may be added to beverages, solid food, semi-solid food, and other consumable substances in the any food composition.
  • Additives, fillers, and excipients may include acidulents, acidity regulators, anticaking agents, antifoaming and foaming agents, antioxidants, bulking agents, food coloring, color retention agents, emulsifiers, flavors, flavor enhancers, four treatment agents, glazing agents, humectants, tracer gas, preservatives, stabilizers, sweeteners, and thickeners.
  • additives, fillers and excipients include one or more corn starch, maltodextrin 10, cellulose, mineral mix S10001, sodium bicarbonate, vitamin mix V10001, and choline bitrartrate.
  • the additives are food coloring.
  • Food coloring may include FD&C blue #1, FD&C blue #2, FD&C citrus red #2, FD&C green #3, FD&C red #3, FD&C red #40, FD&C yellow #5, and FD&C yellow #6.
  • dyes in the food composition include one or more FD&C red #40, FD&C blue #1, and FD&C yellow dye #5.
  • the methods further comprises preservatives.
  • Preservatives may be added to beverages, solid food, semi-solid food, and other consumable substances in the food composition.
  • Preservatives may include natural food preservatives, chemical food preservatives, or artificial preservatives.
  • preservatives may include acetic acid, ascorbic acid, calcium ascorbate, erythrobic acid, isoascorbic acid, potassium nitrate, potassium nitrite, sodium ascorbate, sodium erythorbate, sodium isoascorbate, sodium nitrate, sodium nitrite, wood smoke, benzoic acid, calcium sorbate, carnobacterium divergens M35 , carnobacterium maltaromaticum CB1, ethyl lauroyl arginate, 4-hexylresorcinol, leuconostoc carnosum 4010, methyl-p-hydroxy benzoate, methyl paraben, potassium acetate, potassium benzoate, potassium bisulphite, potassium diacetate, potassium lactate, potassium metabisulphite, potassium sorbate, propyl-p-hydroxy benzoate, propyl paraben, sodium acetate, sodium benzoate, sodium bisulphite, sodium diacetate, sodium lactate
  • preservatives are selected from a group consisting of sulfites, vitamin E, vitamin C, and butylated hydroxytoluene.
  • preservation of beverages, solid food, semi-solid food, and other consumable substances may be accomplished through physical preservation, such as through freezing, boiling, smoking, and dehydration.
  • the liquid composition is stored in a container.
  • the container may be a dialysis bag.
  • the dialysis bag is intended for single-use.
  • the container may hold about 1500 to 5000 mL of liquid composition. In one embodiment, approximately 2000 mL of liquid composition is a single unit. In another embodiment, approximately 2500 mL of liquid composition is a single unit.
  • the dry composition or concentrated composition is stored in a container.
  • the container may be a dialysis bag.
  • the dialysis bag is intended for single-use.
  • the dry composition or concentrated composition further comprises an aqueous solution in the container, thereby producing a mixture.
  • the aqueous solution may be a sterile or nonsterile fluid.
  • the container comprises approximately 2000 mL of aqueous solution.
  • the container comprises approximately 2500 mL of aqueous solution.
  • the mixture has a pH of approximately 4.0-6.5.
  • the mixture has an osmolarity of approximately 300-720 mOsmol/L.
  • the present disclosure provides a method for treating cancer and preventing cancer with the food compositions and/or dialysis compositions disclosed herein.
  • the method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g., Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CLL), chronic myelogenous leukemia (CLL), chronic mye
  • the cancer is selected from a group consisting of leukemia, hematologic malignancy, solid tumor cancer, prostate cancer, breast cancer, liver cancer, or brain tumor.
  • the cancer is selected from a group consisting of lung cancer, liver cancer, breast cancer, prostate cancer, colon cancer, lymphoma, and leukemia.
  • the present disclosure provides a method of treating cancer in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • the present disclosure provides a method of depleting cysteine in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • the dialysis comprises administering a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein.
  • the low-cysteine diet is a food composition disclosed herein.
  • the present disclosure provides a method for treating obesity and preventing obesity.
  • the method relates to the treatment or prevention of disorders that may result in obesity or be the cause of obesity such as overeating and bulimia, polycystic ovarian disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, Type II diabetics, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g., children with acute lymphoblastic leukemia.
  • the method relates to treating at least one obesity-related health condition, including, not limited to, cardiovascular diseases, type 2 diabetes, obstructive sleep apnea, cancer, osteoarthritis, and depression.
  • the present disclosure provides a method for treating renal disease and preventing renal disease.
  • renal disease is chronic renal disease.
  • the method relates to treating a subject diagnosed with renal disorders as identified by medical history, physical findings and laboratory tests.
  • the subject may be diagnosed with acute or chronic renal failure, resulting from diabetes, ischemia, drug induced toxicity, post-transplantation rejection with or without the need for dialysis; glomerulonephritis; glomerulosclerosis; interstitial nephritis; and acute tubular necrosis.
  • Subjects may have physical findings such as anuria, lethargy, coma and decreased general growth rate.
  • Subject may have increased plasma levels of creatinine, urea and uric acid (BUN), proteinuria, decreased GFR, RPF and renal size as determined by urogram, altered acid/base balance and changes in urine specific gravity.
  • BUN creatinine, urea and uric acid
  • the subject is a human.
  • the human subject may be an adult, a child, or an infant.
  • the human subject is a human adult.
  • the human subject may be a human adult 18 years old or older.
  • the human subject may be between the ages 18 and 90 years old.
  • the human subject is between 40 and 85 years old.
  • the human subject is between 65 and 80 years old.
  • the subject is an animal.
  • the animal subject may be a domesticated animal, such as a dog or cat.
  • the animal subject may be a mouse or rat.
  • the animal subject may be a mouse or rat at least 7 weeks old.
  • the animal subject may be a mouse or rat between 7 and 15 weeks old.
  • the animal subject may be a mouse or rat between 10 and 11 weeks old.
  • Subjects that can be treated with a food composition, liquid composition, concentrated composition, and/or dry composition of the present disclosure include subjects that have been diagnosed as having acute myeloid leukemia, acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g., Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CIVIL), chronic myleoproliferative disorders colon cancer, colorectal
  • the subjects treated with a food composition, liquid composition, and/or dry composition of the present disclosure include subjects that have been diagnosed as having a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, and prostate (e.g., benign prostatic hypertrophy (BPH).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, and prostate (e.g., benign prostatic hypertrophy (BPH).
  • the human subject has been diagnosed with leukemia, lung cancer, liver cancer, breast cancer, colon cancer, prostate cancer, stomach cancer, cervix uteri, esophagus, bladder, non-hodgkin lymphoma, and combinations thereof.
  • the animal subject has been diagnosed with malignant lymphoma, skin cancer, mammary gland tumors, breast cancer, soft tissue sarcoma, bone cancer, and combinations thereof.
  • the subject has a genetic predisposition for cancer.
  • the subject has elevated cysteine biosynthetic gene expression levels.
  • the term “obesity” refers to a condition in which individuals are considered “overweight,” or “obese.”
  • the term “obese” or “obesity” includes “extreme obesity,” “severe obesity,” and “super obesity.”
  • the subject is a human.
  • the human subject may be an adult or a child.
  • the human subject is a human child.
  • a human child may be between the ages of 2 and 5 years old, the ages of 6 and 11 years old, and the ages of 12 and 17 years old.
  • the human subject is a human adult.
  • the human subject may be a human adult 18 years old or older.
  • the human subject may be 18-90 years old.
  • the human subject is 30-80 years old.
  • the human subject is 45-65 years old.
  • the subject is an animal.
  • the animal subject may be a domesticated animal, such as a dog or cat.
  • the animal subject may be a mouse or rat.
  • the animal subject may be a mouse or rat at least 7 weeks old.
  • the animal subject may be a mouse or rat that is 7-15 weeks old.
  • the animal subject may be a mouse or rat that is 10-11 weeks old.
  • Subjects that can be treated with a food composition, liquid composition, concentrated composition, and/or dry composition of the present disclosure include subjects that have been diagnosed as being overweight or obese and/or having a body mass index (BMI) of at least 25 kg/m 2 . Obesity in a subject can be assessed using a body mass index (BMI) measurement. A subject's BMI can be calculated by dividing the subject's body weight (in kg) by the square of the subject's height (m 2 ).
  • a subject can be considered obese if the subject exhibits a BMI that is over 25 kg/m 2 , over 26 kg/m 2 , over 27 kg/m 2 , over 28 kg/m 2 , over 29 kg/m 2 , over 30 kg/m 2 , over 31 kg/m 2 , over 32 kg/m 2 , over 33 kg/m 2 , over 34 kg/m 2 , over 35 kg/m 2 , over 36 kg/m 2 , over 37 kg/m 2 , over 38 kg/m 2 , over 39 kg/m 2 , over 40 kg/m 2 , over 41 kg/m 2 , over 42 kg/m 2 , over 43 kg/m 2 , over 44 kg/m 2 , over 45 kg/m 2 , over 46 kg/m 2 , over 47 kg/m 2 , over 48 kg/m 2 , over 49 kg/m 2 , or over 50 kg/m 2 .
  • the subject may be diagnosed with class I obesity, class II obesity, class III obesity, or an equivalent thereof.
  • the subject may have a BMI of at least 30 kg/m 2 , at least 35 kg/m 2 , at least 40 kg/m 2 , at least 45 kg/m 2 or at least 50 kg/m 2 .
  • the subject may have a BMI between 25 kg/m 2 and 50 kg/m 2 .
  • the subject may experience at least one obesity-related health condition, including, not limited to, cardiovascular diseases, type 2 diabetes, obstructive sleep apnea, cancer, osteoarthritis, and depression.
  • a subject can also be considered to exhibit a propensity to develop NASH or hepatic steatosis if the subject exhibits abdominal obesity.
  • Abdominal obesity can be assessed by measuring the circumference of the subject's waist. For example, if the subject is an adult male, the subject can be considered to exhibit abdominal obesity if the subject exhibits a waist circumference of 102 cm or greater. For other example, if the subject is an adult female, the subject can be considered to exhibit abdominal obesity if the subject exhibits a waist circumference of 88 cm or greater.
  • the subject habitually consumes an excessive amount of food, has a genetic predisposition for obesity, routinely lacks physical activity, has an adverse reaction to medication, has a mental disorder, has an endocrine disorder, and combinations thereof.
  • the subject has a genetic predisposition for obesity.
  • obesity in the subject is due to polymorphisms in one or more genes.
  • the subject habitually consumes an excessive amount of food.
  • treating obesity is characterized by the subject losing at least 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 10% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 20% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 30% of the subject's initial body weight.
  • kidney disease includes, but is not limited to, kidney disease, nephropathy, nephritis, nephrosis, chronic kidney disease, chronic renal failure, acute renal disease and acute kidney injury.
  • the subject is a human.
  • the human subject may be an adult or a child.
  • the human subject is a human child.
  • the human subject is a human adult.
  • the human subject may be a human adult 18 years old or older.
  • the human subject is at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 years old.
  • the human subject may be at least 60 years old.
  • the human subject may be between the ages 18 and 90 years old. In an exemplary embodiment, the human subject is between 55 and 75 years old.
  • the subject is an animal.
  • the animal subject may be a domesticated animal, such as a dog or cat.
  • the animal subject may be a mouse or rat.
  • the animal subject may be a mouse or rat at least 7 weeks old.
  • the animal subject may be a mouse or rat between 7 and 15 weeks old.
  • the animal subject may be a mouse or rat that is 10-11 weeks old.
  • Subjects that can be treated with a food composition, liquid composition, and/or dry composition of the present disclosure include subjects that have been diagnosed with renal disease and/or a related condition.
  • the subject has been previously or concurrently diagnosed with high blood pressure and/or diabetes, has an adverse reaction to medication, has a genetic predisposition for renal disease, experiences blockage to the urinary system, experiences inflammation (such as glomerulonephritis), and/or experiences infections (such as pyelonephritis).
  • the present disclosure provides a facility comprising a plurality of liquid compositions.
  • the facility manufactures the liquid composition described herein or components thereof.
  • the facility is a pharmaceutical manufacturing site.
  • the facility distributes the liquid composition described herein or components thereof.
  • the facility may be a distribution center.
  • the facility administers the liquid composition described herein or components thereof.
  • the facility is a hospital, dialysis center, home or clinic. The facility may administer the liquid composition to a subject through dialysis.
  • the present disclosure provides a facility comprising a plurality of dry compositions or concentrated compositions.
  • the facility manufactures the dry composition or concentrated composition described herein or components thereof.
  • the facility is a pharmaceutical manufacturing site.
  • the facility distributes the dry composition or concentrated composition described herein or components thereof.
  • the facility may be a distribution center.
  • the facility administers the dry composition or concentrated composition described herein or components thereof.
  • the facility is a hospital or clinic. The facility may administer the dry composition or concentrated composition to a subject through dialysis.
  • the present disclosure provides a facility comprising a plurality of food compositions.
  • the facility manufactures the food compositions described herein or components thereof.
  • the facility is a food manufacturing site.
  • the facility distributes the food compositions described herein or components thereof.
  • the facility may be a distribution center.
  • the facility provides the food compositions described herein and components thereof to the subject.
  • the facility may sell the food composition to the subject.
  • the facility may prepare the food composition.
  • the facility may administer the food composition to the subject.
  • the facility may be a distribution center, a commercial kitchen, a restaurant, an eating facility, or a medical facility.
  • the facility is a location that provides catering or food delivery service to hospitals, clinic or patients. The facility may be located in or within the vicinity of a hospital or clinic, such as within 0.1, 0.5, 1, or 5 miles of a hospital or clinic.
  • kits comprising: (a) a pre-packaged food composition as described herein; and (b) instructions for using the pre-packaged food composition.
  • the kits include one or more food compositions described herein, in suitable packaging, and can further comprise written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like.
  • the instructions may comprise an informational package describing the use and attendant benefits of the food composition in treating cancer, obesity or renal disease.
  • kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the food composition, and/or which describe dosing, administration, the dieting regimen, side effects, drug interactions, or other information useful to the health care provider. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
  • a kit can comprise one or more complete meals and/or snacks of the food composition described herein.
  • a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more complete meals and/or snacks of the food composition.
  • the kits may provide complete meals and/or snacks for exactly or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, or more days.
  • Instructions for use can comprise diet regimen instructions, such as instructions to take 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more complete meals and/or snacks 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times per day.
  • kits can comprise a complete meal and/or snack of the food composition supplied as a food product, with each food product packaged separately, multiples of a food product packaged separately according to the number of complete meals and/or snacks of the food composition per administration (e.g. pairs of components), or all food products packaged together (e.g. in a container).
  • a kit can comprise a complete meal and/or snack of the food composition as a bottled drink, the kit comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 24, 28, 36, 48, 72, or more bottles.
  • kits can comprise a complete meal and/or snack of the food composition as a nutritional bar, the kit comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 24, 28, 36, 48, 72, or more nutritional bar.
  • the kit may comprise components of the food composition provided or packaged as separate compositions in separate containers within the kit.
  • the kit may comprise components of the food composition provided or packaged as a single composition within a container of the kit.
  • the kit can further contain another agent.
  • the food composition or component thereof, and the agent are provided or packaged as separate compositions in separate containers within the kit.
  • the food composition or component thereof, and the agent are provided or packaged as a single composition within a container in the kit.
  • Suitable packaging and additional articles for use e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like
  • Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits can also, in some embodiments, be marketed directly to the consumer.
  • the kit can further contain a pre-packaged liquid composition, pre-packaged concentrated composition, or a pre-packaged dry composition described herein.
  • the kit can comprise instructions directing the administration of the pre-packaged liquid composition, or pre-packaged concentrated composition, or dry composition.
  • a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more pre-packaged liquid compositions, pre-packaged concentrated compositions or pre-packaged dry compositions.
  • a kit can comprise a multi-day supply of complete meals and/or snacks of the food composition as described herein.
  • the kit can comprise instructions directing the administration of the multi-day supply of complete meals and/or snacks of the food composition over a period of multiple days.
  • the multi-day supply can be a one-month supply, a two-month supply, or a multi-week supply.
  • the multi-day supply can be a 90-day, 180-day, 3-month or 6-month supply.
  • the kit can include packaged daily complete meals and/or snacks of the food composition, such as packages of 1, 2, 3, 4, or 5 complete meals and/or snacks of the food composition.
  • the kit can be packaged with other dietary supplements, vitamins, and/or meal replacement bars, mixes, and beverages.
  • the present disclosure provides a kit comprising: (a) a pre-packaged liquid composition as described herein; and (b) instructions for using the pre-packaged liquid composition.
  • the present disclosure provides a kit comprising: (a) a pre-packaged dry composition or concentrated composition as described herein; and (b) instructions for using the pre-packaged dry composition.
  • the kits include one or more liquid compositions, concentrated compositions, or dry compositions described herein, in suitable packaging, and can further comprise written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like.
  • the instructions may comprise an informational package describing the use and attendant benefits of the liquid composition or dry composition in treating cancer, obesity or renal disease.
  • kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the liquid composition, concentrated composition, or dry composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
  • a kit can comprise one or more dialysis bags containing the liquid composition, concentrated composition, or dry composition described herein.
  • a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more dialysis bags containing the liquid composition, concentrated composition, or dry composition.
  • the kits may provide dialysis bags containing the liquid composition, concentrated composition, or dry composition for exactly or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, or more days.
  • Instructions for use can comprise dialysis regimen instructions, such as instructions to use 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more dialysis bags containing the liquid composition, concentrated composition, or dry composition 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times per day.
  • a kit can comprise a dialysis bag containing the liquid composition, concentrated composition, or dry composition supplied as a single-use dialysis bag, with each single-use dialysis bag packaged separately, multiples of a single-use dialysis bag packaged separately according to the number of dialysis bags containing the liquid composition, concentrated composition, or dry composition per administration (e.g. pairs of single-use dialysis bag), or all single-use dialysis bag packaged together (e.g. in a container).
  • the kit may comprise components of the dialysis bag containing the liquid composition, concentrated composition, or dry composition provided or packaged as separate compositions in separate containers within the kit.
  • the kit may comprise components of the dialysis bag containing the liquid composition, concentrated composition, or dry composition provided or packaged as a single composition within a container of the kit.
  • the kit can further contain another agent.
  • the dialysis bag containing the liquid composition, concentrated composition, or dry composition or component thereof, and the agent are provided or packaged as separate compositions in separate containers within the kit.
  • the liquid composition, concentrated composition, dry composition, or component thereof, and the agent are provided or packaged as a single composition within a container in the kit.
  • Suitable packaging and additional articles for use e.g., aqueous solution for reconstituting the dry composition or concentrated composition, measuring cup for liquid preparations, wrapping to minimize exposure to air, and the like
  • Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits can also, in some embodiments, be marketed directly to the consumer.
  • the kit can further contain a pre-packaged liquid composition, pre-packaged concentrated composition, or a pre-packaged dry composition described herein.
  • the kit can comprise instructions directing the administration of the pre-packaged liquid composition, pre-packaged concentrated composition or pre-packaged dry composition.
  • a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more pre-packaged liquid compositions, pre-packaged concentrated compositions or pre-packaged dry compositions.
  • a kit can comprise a multi-day supply of dialysis bags containing the liquid composition, concentrated composition, or dry composition as described herein.
  • the kit can comprise instructions directing the administration of the multi-day supply of dialysis bags containing the liquid composition, concentrated composition, or dry composition over a period of multiple days.
  • the multi-day supply can be a one-month supply, a two-month supply, or a multi-week supply.
  • the multi-day supply can be a 90-day, 180-day, 3-month or 6-month supply.
  • the kit can include packaged dialysis bags containing the liquid composition, concentrated composition, or dry composition, such as packages of 1, 2, 3, 4, or 5 dialysis bags containing the liquid composition, concentrated composition, or dry composition.
  • the kit can be packaged with a food composition described herein, dietary supplements, vitamins, and/or meal replacement bars, mixes, and beverages.
  • the present disclosure provides a system for placing a subject under dialysis and/or a nutritional diet with a composition disclosed herein to aid in the treatment or prevention of a disease.
  • the system may carry out any of the methods disclosed herein and may use any of the compositions disclosed herein.
  • the dialysis systems of the present disclosure can be controlled and modified to provide various dialysis therapies, as desired.
  • the dialysis systems may be modified (e.g. increase/decrease the number of exchanges, increase/decrease the amount of dialysate used during each exchange, and/or use an adjusted dialysate formulation) to adjust to a subject's medical progress.
  • the dialysis systems may facilitate peritoneal dialysis, hemodialysis and other forms of dialysis.
  • the dialysis systems may comprise (a) a dialysis container comprising a composition disclosed herein, (b) tubing, and (c) a collection container.
  • the dialysis systems further comprise a machine for facilitating and/or automating fluid exchange.
  • the present disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein, and a dialysis machine configured to administer the liquid composition, the concentrated composition or the dry composition.
  • the dialysis machine comprises a pump, a pressure monitor, air trap, air detector, a clamp, and/or a filter.
  • the present disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein.
  • Peritoneal dialysis is a treatment that uses the lining of a subject's abdomen, i.e. the peritoneum, and a dialysate to clean the subject's blood. Any of the liquid compositions, concentrated compositions, and dry compositions may be used as a dialysate in PD. Dialysate absorbs waste and fluid from the blood, using the peritoneum as a filter.
  • the dialysis system involves continuous ambulatory PD (CAPD) or continuous cycler-assisted PD (CCPD).
  • CAPD continuous ambulatory PD
  • CCPD continuous cycler-assisted PD
  • CAPD is “continuous,” machine-free and can be done at any time.
  • the treatment may involve placing about two quarts of cleansing fluid into the subject's abdomen and later draining it.
  • CAPD can be done by hooking up a plastic bag of cleansing fluid to the tube in the peritoneum.
  • the subject may raise the dialysis bag to approximately shoulder level to pull the dialysis fluid into the peritoneum.
  • the dialysis bag is removed and discarded.
  • an exchange putting in and taking out the fluid
  • the fluid is drained from the subject's abdomen. In some embodiments, this process is performed several times in a 24-hour period, such as 1, 2, 3, 4, 5, 6 or 7 times, or as needed.
  • FIG. 14A shows an exemplary CAPD process (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/peritoneal-dialysis).
  • a patient undergoing CAPD may hooked up to a dialysis bag 101 and waste container 105 by a catheter placed in the patient's peritoneum and connecting tubes 104 .
  • the transfer set 103 connects the catheter to the dialysis bag 101 to perform dialysis exchange and can be designed to reduce the risk of infection.
  • the transfer set may be “Y-shaped,” and one branch of the Y-tube may connect to the waste container 105 , while the other branch may connect to a dialysis bag 101 containing a dialysis solution as described herein.
  • the dialysis solution is warmed prior to use by the subject.
  • the dialysis bag 101 is placed above the patient's peritoneum in a way to allow the fluid in the dialysis bag to enter into the patient, such as on a pole as illustrated in FIG. 14A .
  • CCPD also known as automated peritoneal dialysis (APD)
  • APD automated peritoneal dialysis
  • the treatment may be performed at any time, especially at night.
  • the dialysis system automatically performs dialysis therapy on a patient, for example, during nighttime while the patient sleeps.
  • the subject may be attached to the machine for an extended period of time, such as 8, 9, 10, 11, or 12 hours per day.
  • FIG. 14B shows an exemplary CCPD process (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/peritoneal-dialysis). As shown in FIG.
  • a patient undergoing CCPD may hooked up to a supply of dialysis solution disclosed herein 201 and waste line 205 .
  • the dialysis may be facilitated by a dialysis machine 204 (also known as a cycler).
  • the dialysis machine may further comprise a heater 203 .
  • the flow of dialysis solution may be monitored by a fluid meter 202 .
  • the fluid meter 202 may measure and record how much solution the cycler 204 removes.
  • the cycler 204 compares the amount of dialysis solution that was put into the subject with the amount of fluid that drains out.
  • the cycler 204 may to fill and empty the subject's peritoneum several times a night, such as three to five times, while the subject sleeps.
  • Hemodialysis is a treatment to filter wastes and water from your blood.
  • a dialysis machine and a special filter called an “artificial kidney,” or a “dialyzer,” are used to clean a subject's blood.
  • the blood goes through the dialyzer, which is located outside of the subject's body.
  • FIG. 15 is an exemplary schematic representation of hemodialysis (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/hemodialysis).
  • FIG. 15 is an exemplary schematic representation of hemodialysis (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/hemodialysis).
  • hemodialysis provides an overview of the filtering process in hemodialysis (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/hemodialysis).
  • a patient undergoing hemodialysis may be connected to a dialysis machine by inserting two needles in the subject's arm. Each needle is attached to a soft tube connected to the dialysis machine. The dialysis machine pumps blood through the filter and returns the blood to the patient. Through one of the needles, blood is removed for dialysis 307 .
  • the unfiltered and filtered blood is monitored with an arterial pressure monitor 308 , dialyzer inflow pressure monitor 302 , and venous pressure monitor 304 .
  • An anticoagulant 301 may be used to prevent blood clotting.
  • the dialysis machine pumps the blood through a filter 303 (also known as a dialyzer) with a pump 309 .
  • the dialyzer 303 has two parts, one for your blood and one for a washing fluid called dialysate. A thin membrane separates these two parts.
  • dialysis solution 404 as disclosed herein passes in the opposite direction on the outside of the fibers. Waste products from the blood move into the dialysis solution and out of the filter 403 . Filtered blood remains in the hollow fibers and returns to the body. Some nutrients from the dialysis solution may also return to the body with the filtered blood.
  • hemodialysis is performed about 1, 2, 3, 4, 5, 6 or 7 times per week for about 1, 2, 3, 4, 5 or 6 hours at a time, or as needed.
  • a cancer therapy regimen is provided to a patient previously identified as having a form of cancer.
  • the patient is diagnosed with a tumor, solid or liquid, in the primary or metastatic stage.
  • the regimen involves utilizing a combination of a nutritionally modified diet and dialysis with compositions disclosed here.
  • the regimen involves metabolic intervention therapy that can be tailored to the needs of the individual patient.
  • a metabolic cancer therapy can be provided to a patient diagnosed with cancer.
  • the cancer patient is treated with dialysis and/or a nutritional diet in order to deplete targeted nutrient(s)
  • the patient is primed with any food composition described herein, i.e. a depleting diet, for approximately 3 days (“the priming stage”).
  • the patient's entire diet comprises any food composition described herein, with the exception of food and beverages approved by the regimen.
  • the patient's plasma amino acid levels are measured and profiled by targeted LC-MS assay before and after the priming stage.
  • dialysis treatment e.g. hemodialysis or peritoneal dialysis.
  • the patient may undergo hemodialysis or peritoneal dialysis alone, or undergo either form dialysis serially or interchangeably within the therapy.
  • a patient undergoing hemodialysis undergoes dialysis with a liquid composition, concentrated composition, or dry composition described herein (“dialysis solution”) at a facility described herein.
  • the patient receives the dialysis solution for approximately 4 hours during the day or overnight.
  • the patient receives dialysis treatment at least 3 times a week, for approximately 4 hours for each dialysis treatment.
  • the patient's plasma amino acid profile is analyzed for monitoring treatment effect progression.
  • a patient undergoing peritoneal dialysis undergoes dialysis with a liquid composition, concentrated composition, or dry composition described herein (“dialysis solution”) at a facility described herein, at the patient's home, or a convenient location.
  • the patient receives the dialysis solution for approximately 8-12 hours per day or 24 hours continuously.
  • the patient carries a portable device that administers the dialysis solution.
  • the patient's plasma amino acid profile is analyzed approximately every 3 days for monitoring treatment effect progression.
  • the patient's amino acid profile achieves therapeutic depletion levels after no more than approximately 8 weeks, as determined by the LC-MS assay. At this point, the patient discontinues dialysis treatment, but continues to take depleting diets until complete remission is achieved. Once the patient reaches complete remission, the patient remains on the depleting diet for approximately 2 weeks to stabilize complete remission, in which the patient is tested throughout.
  • the patient's disease progression is monitored by companion test. Any serious complications that arise may be conveniently stopped or limited by allowing the patient to resume a normal diet.
  • the cancer therapy regimen is resumed once the patient's condition is stabilized. The patient is permitted to continue to take painkillers or other cancer medicines as long as it does not interfere with the metabolic intervention treatment.
  • dietary intervention may be administrated for 1-4 weeks with companion plasma nutrient test to indicate the safety and potential efficacy in an individual patient.
  • companion plasma nutrient test As described in Example 1, during the test, the patient will only eat and drink the prescribed food composition.
  • the diet regimen may be administrated as long as the subject remains obese.
  • the renal disease patient may solely rely on dialysis for normal nutrient nourishment through the administration of the dry composition, concentrated composition, or liquid composition described herein.
  • the dialysis procedure as set forth in Example 1, is followed.
  • dialysis may be administrated as long as the subject's renal function remains impaired or lost.
  • dietary intervention may be administrated for 1-4 weeks with companion cancer progression test and plasma nutrient test to indicate the safety and potential efficacy in an individual patient.
  • companion cancer progression test and plasma nutrient test As described in Example 1, during the test, the patient will only eat and drink the prescribed food.
  • the patient on the diet regimen may be administrated to the subject as long as the tumor is not completely cured.
  • the cancer patient may solely rely on dialysis both for targeted depletion of cancer-addicted nutrients in plasma and for normal nutrient nourishment through the administration of the dry composition, concentrated composition, or liquid composition described herein.
  • a dialysis only treatment is particularly applicable to late-stage gastrointestinal cancer because patients usually lose the ability to eat and rely solely on nutrient infusion.
  • dialysis may be administrated to the subject as long as the tumor is not completely cured.
  • Table 2 as presented below provides an exemplary formulation of the dialysis fluid—dry composition, concentrated composition, or liquid composition—described herein.
  • Dialysis fluid composition Dextrose (/L) mmole Hemodialysis only 5.5-11.0 peritoneal dialysis only 83-236 Osmolarity (mOsmol/L) 300-718 pH 6.0 (4.0-6.5) mmole mEq Ionic Concentration (/L) Sodium 132 132 Calcium 1.75 3.5 Potassium 2 2 Magnesium 0.25 0.5 Chloride 96 96 Buffer (L) HCO3 20-50 20-50 Acetate 2.0-5.0 2.0-5.0 Lactate 40 40 Amino acid (/L) micromole MW microgram Alanine 347.5 89.0935 30.96 Arginine 60.25 174.2017 10.50 Asparagine 57.5 132.1184 7.60 Aspartic acid 8 133.1032 1.06 Beta-alanine 19.5 89.0935 1.74 Cystine 40.25 240.3 9.67 Glutamic acid 72 147.1299 10.59 Glutamine 547.5 146.1451 80.01 Glycine 212.5 75.0669
  • Table 3 as presented below provides an exemplary formulation of the food composition described herein.
  • FIG. 3 shows an overview of the diet compositions used in the studies of the present disclosure and Table 5 of Example 14 further describes the diets.
  • the “complete diet” was the “designed control” of FIG. 3 .
  • MEAD201 and MEAD203 were depleted in 5 and 9 non-essential amino acids, respectively.
  • MEAD202 was lacking 3 non-essential amino acids and insufficient 1 non-essential amino acid.
  • Protein Y is natural protein casein, a common food ingredient to provide protein.
  • Protein Z is soy protein.
  • Protein X is collagen (gelatin).
  • mice were implanted with HCT116 cells by subcutaneous injection. The mice were approximately 5-6 week old. The study had two groups: mice administered with a complete diet and mice administered with MEAD202. Each group had 5 mice. The mice were placed on the respective diet on day 7. The mice were fed ad libitum. The complete diet and MEAD 202 compositions are described in Table 5 of Example 14. The tumor sizes were monitored 3 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • mice that were treated with MEAD202 exhibited a significant decrease in tumor volume in comparison to the mice that were treated with a complete diet.
  • the mice treated with MEAD202 consistently had a lower tumor volume than the mice treated the complete diet.
  • the tumor volume for the MEAD202 mice were over 25% lower than the complete diet mice.
  • mice NOD-SCID mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study in FIG. 5A had two groups: mice administered with a complete diet and mice administered with treatment diets [MEAD202 (day 28-45) and MEAD203 (after day 45)], and the mice were placed on the respective diet on day 20.
  • athymic nude mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study in FIG. 5B had 3 groups: mice administered with a complete diet, mice administered with MEAD202, and mice administered with MEAD203, and the mice were place on the respective diet on day 7. Each group had 5 mice.
  • mice were fed ad libitum.
  • the tumor sizes were monitored 3 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • mice that were treated with a nutritional diet exhibited a significant decrease in tumor volume than the mice treated with a complete diet.
  • the mice treated with the treatment diets consistently had a lower tumor volume than the mice treated the complete diet.
  • the tumor volume for the treatment diets mice were approximately 60% lower than the complete diet mice.
  • the mice treated with MEAD203 and MEAD202 consistently had a lower tumor volume than the mice treated the complete diet.
  • the tumor volume for both the MEAD203 and MEAD202 mice were over 25% lower than the complete diet mice.
  • mice were implanted with PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had two groups: mice administered with a complete diet and mice administered with MEAD203. Each group had 5 mice. The mice were placed on the respective diet treatment on day 5. The mice were fed ad libitum. The complete diet and MEAD 203 compositions are described in Table 5 of Example 14. The tumor sizes were monitored 3 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • mice that were treated with MEAD203 exhibited a significant decrease in tumor volume in comparison to the mice that were treated with a complete diet.
  • the mice treated with MEAD203 consistently had a lower tumor volume than the mice treated the complete diet.
  • the tumor volume for the MEAD203 mice were approximately 60% lower than the complete diet mice.
  • mice were implanted with MIAPaCa-2 cells by subcutaneous injection. The mice were 6-8 weeks old. The study had two groups: mice administered with a complete diet and mice administered with treatment diets [MEAD201 (day 0-24), MEAD202 (day 25-39) and MEAD203 (after day 39)]. Each group had 5 mice. The mice were placed on the respective diet on day 0. The mice were implanted 7 days before being placed on the respective diet. The complete diet and MEAD 202 compositions are described in Table 5 of Example 14. The mice were fed ad libitum. The tumor sizes were monitored 2 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • mice that were treated with the treatment diets exhibited a significant decrease in tumor volume in comparison to the mice that were treated with a complete diet.
  • the mice treated with the treatment diets on average had a lower tumor volume than the mice treated the complete diet.
  • the tumor volume for the treatment diet mice were over 40% lower than the complete diet mice.
  • MEAD102 MEAD102
  • MEAD103 MEAD1 ⁇ M
  • an 8-component vitamin cocktail 10-component vitamin cocktail
  • 10-component vitamin cocktail 10-component vitamin cocktail
  • 10-component vitamin cocktail 10-component vitamin cocktail
  • 10-component vitamin cocktail 10-component vitamin cocktail
  • 10-component vitamin cocktail 10-component vitamin cocktail
  • MEAD102 the vitamin cocktail comprised: vitamin B6, folate (Vit B9), thiamine, riboflavin, vitamin B12, biotin, choline, and inositol.
  • MEAD103 the vitamin cocktail comprised the MEAD102 cocktail, and niacinamide and pantothenic-Calcium.
  • the compositions for MEAD101, MEAD102, MEAD103 and MEAD104 are described below in Table 8.
  • mice were implanted with U87MG cells by subcutaneous injection. The mice were 6-8 weeks old.
  • the study in FIG. 9 had 3 groups: mice administered with a complete diet, mice administered with MEAD203, and mice administered with MEAD 203 and placed on dialysis with MEAD101, MEAD102 and MEAD103 at various time points. Each treatment group had 1 mouse. The mice were placed on the respective treatment on day 5. The mice were implanted 7 days before being placed on the respective diet. The mice were fed ad libitum. For dialysis session, approximately 2 mL of dialysis solution was injected in the peritoneal space, and drawn out approximately 2 hours later. The tumor sizes were monitored daily. Arrows indicate the day and type of dialysis fluid were used.
  • mice administered a complete diet mice administered with diet only, mice administered with dialysis only, and mice administered with diet and dialysis. Each group had 6 mice. The mice were placed on the respective treatment on day 1. Diet MEAD203 was used as the treatment diet. Designed Control was used as the control diet. The mice were implanted 22 days before being placed on the respective diets. The mice were fed ad libitum. MEAD103 was used for the dialysis. For each dialysis session, approximately 2 mL of fluid was injected in the peritoneal space, and drawn out approximately 2 hours later. Dialysis was performed on every week day. Mice were monitored every weekday.
  • mice on the MEAD203 diet and dialysis experienced a reduction in tumor volume.
  • the mice on the complete diet were euthanized at day 16 because the tumor volume was over 2000 mm3 for 3 consecutive measurements. Additionally, the mice were euthanized due to concerns of over ulceration and infection between mice.
  • the mice that were administered a treatment of diet+dialysis and the mice treated with diet only exhibited a reduction in tumor volume as compared to the mice on a complete diet and the mice on dialysis only.
  • the diet+dialysis mice consistently had the lowest tumor volume in comparison to the other treatment groups.
  • the mice that were administered a treatment of diet+dialysis and the mice treated with diet only exhibited approximately a 40% reduction in tumor volume as compared to the mice on a complete diet and the mice on dialysis only.
  • mice were implanted with MDA-MDB-231 cells by subcutaneous injection.
  • the mice were 5-6 weeks old.
  • the study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in Example 16.
  • Each group had 5 mice.
  • the mice were placed on respective diets on day 20.
  • the complete diet and treatment diet compositions are described in Table 5 of Example 14.
  • the mice were fed ad libitum.
  • the body weights were monitored 3 times weekly. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • mice were implanted with PC-3 cells by subcutaneous injection. mice that were implanted with implanted PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 2 groups: mice administered with MEAD203, and mice administered with complete diet. Each group had 5 mice. The mice were placed on respective diets on day 5. The mice were fed ad libitum. The complete diet and MEAD 202 compositions are described in Table 5 of Example 14. The body weights were monitored 3 times weekly. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • mice that were administered a treatment of MEAD203 and the mice treated with MEAD202 exhibited a reduction in body weight as compared to the mice on a complete diet and the mice on natural chow.
  • the mice that were administered MEAD203 and the mice treated with MEAD202 exhibited approximately a 15% reduction in body weight as compared to the mice on a complete diet and the mice on dialysis only. Body weight loss were relatively stabilize for the treatment diets.
  • mice were implanted with MDA-MDB-231 cells by subcutaneous injection.
  • the mice were 5-6 weeks old.
  • the study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in Example 16.
  • Each group had 5 mice.
  • the mice were placed on the respective diet on day 20.
  • the complete diet and treatment diet compositions are described in Table 5 of Example 14.
  • the mice were fed ad libitum.
  • the plasma samples were collected before treatment started and after treatment completed. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001.
  • FIG. 12A athymic nude mice were implanted with MDA-MDB-231 cells by subcutaneous injection.
  • the mice were 5-6 weeks old.
  • the study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in
  • mice 12B athymic nude mice were implanted with PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 2 groups: mice administered with MEAD203, and mice administered with complete diet. Each group had 5 mice. The mice were placed on the respective diet on day 5. The mice were fed ad libitum. The complete diet and MEAD 203 compositions are described in Table 5 of Example 14. The plasma samples were collected before treatment started and after treatment completed. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001. Glucose in plasma was measured with Nova Max Glucose Test Strips.
  • results As shown in FIG. 12A and FIG. 12B , blood sugar is not increased in the treatment groups.
  • the post-dose glucose levels of the MEAD202 mice and MED203 mice were similar or lower than the pre-dose levels.
  • the post-dose glucose levels of the MEAD203 mice were similar to its pre-dose levels.
  • mice were implanted with MDA-MDB-231 cells by subcutaneous injection.
  • the mice were 5-6 weeks old.
  • the study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in Example 16.
  • Each group had 5 mice.
  • the mice were placed on the respective diet on day 20.
  • the complete diet and treatment diet compositions are described in Table 5 of Example 14.
  • the mice were fed ad libitum.
  • the plasma samples were collected before treatment started and after treatment completed. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement.
  • mice athymic nude mice were implanted with PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 2 groups: mice administered with MEAD203, and mice administered with complete diet. Each group had 5 mice. The mice were placed on the respective diet on day 5. The mice were placed on the respective diets 5 days after implantation. The mice were fed ad libitum. The complete diet and MEAD 203 compositions are described in Table 5 of Example 14. The plasma samples were collected before treatment started and after treatment completed. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P ⁇ 0.05, ** for P ⁇ 0.01, *** for P ⁇ 0.001. Beta-ketone body in plasma was measured with Nova Max Ketone Test Strips.
  • ketosis is not triggered by the administration of the treatment diets.
  • the post-dose ⁇ -ketone levels of the MEAD202 mice and MED203 mice were similar to the pre-dose levels.
  • the post-dose ⁇ -ketone levels of the MEAD203 mice were similar to its pre-dose levels.
  • Table 4 as presented below provides an exemplary formulation of the dialysis fluid—dry composition, concentrated composition, or liquid composition—described herein.
  • Table 5 as presented below provides an exemplary formulation of a food composition described herein.
  • the food compositions below were the diets used in studies above.
  • plasma glutamate was increased by about 80-100% in mice when on MEAD treatment diets.
  • the increase in plasma glutamate may result in a reduction of cysteine update due to the effects on the xCT transporter. This could theoretically halve all cells' capability of cysteine uptake.
  • Vitamins have an important function in the reduction of cysteine.
  • Vitamins (especially vitamin B family) are important for dialysis and the nutritional diet disclosed herein.
  • the vitamins may maintain appropriate physiology in normal cells. Vitamins are important for neural health and metabolism. Vitamins may also help normal cells synthesize the target nutrients when these nutrients are not provided through diet or depleted through dialysis.
  • Vitamin C was not included for mouse studies because vitamin C (ascorbic acid) is not vitamin for mice because they can make it internatively. Vitamin C is expected to be important in treating humans. Table 6 below provides a few exemplary vitamins for use in the compositions described herein.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Inorganic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Mycology (AREA)
  • Polymers & Plastics (AREA)
  • Food Science & Technology (AREA)
  • Emergency Medicine (AREA)
  • Nutrition Science (AREA)
  • Anesthesiology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Vascular Medicine (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • External Artificial Organs (AREA)

Abstract

The present disclosure provides compositions and methods for treating cancer with nutritionally modified diets and/or dialysis treatment, such as through amino acid depletion of one or more nonessential amino acids. In some aspects, the present disclosure provides compositions and methods for treating obesity with such modified diets and/or dialysis treatment. Furthermore, in some aspects, the present disclosure provides compositions and methods for treating renal disease with such modified diets and/or dialysis treatment.

Description

    CROSS-REFERENCE
  • This application is a continuation application of U.S. patent application Ser. No. 16/897,152, filed Jun. 9, 2020, which is a continuation application of International Application No. PCT/US2018/065045, filed Dec. 11, 2018, which claims the benefit of U.S. Provisional Application No. 62/597,392, filed on Dec. 11, 2017, each of which is entirely incorporated herein by reference for all purposes.
  • BACKGROUND
  • Cancer and obesity are major public health problems. Cancer is the second leading cause of death globally, and was responsible for 8.8 million deaths in 2012. Globally, approximately 1 in 6 deaths is due to cancer. Cancer arises from the transformation of normal cells into tumor cells in a multistage process that generally progresses from a pre-cancerous lesion to a malignant tumor. One defining feature of cancer is the rapid creation of abnormal cells that grow beyond their usual boundaries, and which can then invade adjoining parts of the body and spread to other organs.
  • Cancer cells adopt a distinctive type of metabolism, the “Warburg effect,” to promote growth, survival, proliferation, and long-term maintenance. This altered metabolism involves uncoupled cellular glycolysis and mitochondrial aerobic respiration, which may lead to the requirement of certain nutrients that are not necessary for normal cells.
  • The currently available treatments suffer from a number of profound drawbacks. For instance, the current cancer treatments available to patients are expensive. The economic impact of cancer is significant and continues to increase. In 2010, the total annual economic cost of cancer was estimated at approximately 1.16 trillion US dollars. Also, the traditional methods of treating cancer, such as chemotherapy and radiation therapy, tend to be highly toxic and/or nonspecific to cancer, which results in limited efficacy and harmful side effects.
  • Obesity is a medical condition in which excess body fat has accumulated to the extent that it may have a negative impact on health. This condition is commonly caused by a combination of excessive food intake, lack of physical activity, and genetic susceptibility. Obesity increases the likelihood of various diseases and conditions, such as, cardiovascular diseases, type 2 diabetes, obstructive sleep apnea, cancer, osteoarthritis and depression.
  • Many of the current treatments for obesity, such as calorie restrictive dietary modification, behavior modification, exercise, medication and surgery face a number of drawbacks. Many obesity treatments are ineffective and provide only short-term results, particularly those who are morbidly obese.
  • Accordingly, there remains a considerable need for new cancer treatments and treatments for obesity.
  • SUMMARY
  • Cancer and metabolic diseases, such as obesity, remain as major health issues in which new, alternative, and effective treatment are currently needed. The present disclosure addresses such needs and provides a number of advantages. The present disclosure relates to providing a subject with a treatment that includes the control of a subject's nutritional diet and/or dialysis. For example, treating a patient in need thereof may comprise placing the patient on a cysteine depleted diet and placing the patient under dialysis. The present disclosure provides solid and liquid compositions that are depleted with one or more nutrients, such as nonessential amino acids, and methods of treating cancer, renal disease, and metabolic diseases, such as obesity, with the compositions. The present disclosure provides for facilities, systems and kits that relate to the distribution and/or performance of the described compositions and methods.
  • The present disclosure utilizes hemodialysis and peritoneal dialysis as a way to deplete one or more nutrients in the subject by administering compositions that are lacking in such nutrients. Hemodialysis involves withdrawing blood from the body and cleaning it in an extracorporeal blood circuit and then returning the cleansed blood to the body. The extracorporeal blood circuit includes a dialyzer which comprises a semipermeable membrane. Within this dialyzer waste substances and excess fluid is removed through the semipermeable membrane, and the semipermeable membrane within the dialyzer has a blood side and a dialysate side. Peritoneal dialysis involves the use of the peritoneum in a person's abdomen as the membrane through which fluid and dissolved substances are exchanged with the blood. Peritoneal dialysis is used to remove excess fluid, correct electrolyte problems, and remove toxins in those with kidney failure. In peritoneal dialysis, a solution is introduced through a permanent tube in the lower abdomen and then removed. This may either occur at regular intervals throughout the day, known as continuous ambulatory dialysis, or at night with the assistance of a machine, known as automated peritoneal dialysis. Furthermore, the present disclosure envisions utilizing hemodialysis and peritoneal dialysis as way to provide a subject in need thereof with a particular nutritional makeup by depleting and/or restoring one or more nutrients in the subject. The present disclosure provides dialysis systems and equipment thereof.
  • In some aspects, the present disclosure provides, a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some aspects, the present disclosure provides a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some aspects, the present disclosure provides a liquid composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (c) at least one essential amino acid; and (d) an aqueous solution, wherein the liquid composition comprises at least 10 μM of essential amino acid, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein the liquid composition has a pH of 4.0-6.5, and wherein the liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some embodiments, the carbohydrate is selected from a group consisting of dextrose and glucose. The carbohydrate may be dextrose. In certain embodiments, the amount of dextrose is 1.0-4.5% w/v. The amount of dextrose may be 1.5-4.25% w/v. The amount of dextrose may be 1.7-4.0% w/v. In another embodiment, the amount of dextrose is 0.05-0.50% w/v. The amount of dextrose may be 0.05-0.25% w/v. The amount of dextrose may be 0.08-0.22% w/v. The amount of dextrose may be 0.1-0.2% w/v.
  • For the liquid composition, in certain embodiments, the liquid composition comprises at least 10 μM of said essential amino acid. In some embodiments, the amount of at least one of the essential amino acid is 10-300 μM. For example, the amount of at least one of the essential amino acid is 20-250 μM. The essential amino acid may be selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine. In some embodiments, at least one of the essential amino acid is methionine. In some embodiments, at least one of the essential amino acid is arginine. The liquid composition may comprise at least two essential amino acids. The liquid composition may comprise at least three essential amino acids. The liquid composition may comprise at least four essential amino acids. The liquid composition may comprise at least five essential amino acids. The liquid composition may comprise at least six essential amino acids. The liquid composition may comprise at least seven essential amino acids. The liquid composition may comprise at least eight essential amino acids. The liquid composition may comprise at least nine essential amino acids. The liquid composition may comprise at least ten essential amino acids.
  • The liquid composition may further comprise at least one vitamin. The vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline, such as for example, the vitamin is vitamin C. In some embodiments, the amount of the vitamin is about 0.000010 to 200 μM. In some embodiments, the liquid composition comprises at least 1 μM of vitamin C. For example, the liquid composition may comprise at least 7 μM of vitamin C. In some embodiments, the vitamin is vitamin B. The liquid composition may comprise 0.000003 to 1500 μM of said vitamin B.
  • The liquid composition may further comprise an essential fatty acid. The essential amino acid may be an omega-3 fatty acid.
  • In some embodiments, the amount of glutamate is approximately 2-500 μM. the amount of glutamate may increase glutamate in the blood by at least 150 μM.
  • In certain embodiments, the liquid composition further comprises at least one mineral. The mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc. The liquid composition may comprise at least 5 nM of the mineral. In some embodiments, the amount of at least one mineral is 5 nM-5 μM.
  • In certain embodiments, the liquid composition is stored in a container. The container may be a dialysis bag. The dialysis bag may be intended for single-use. In some embodiments, the container holds 1500 to 5000 mL of the liquid composition. For example, approximately 2000 mL of the liquid composition is a single unit. In another embodiment, approximately 2500 mL of the liquid composition is a single unit.
  • For the liquid composition, in certain embodiments, the amount of at least one ion is at least approximately 0.1 mEq/L. The amount of at least one ion may be 0.1-150 mEq/L.
  • In some embodiments, liquid composition is free of nonessential amino acids other than glutamate. Alternatively, the liquid composition may further comprise at least one nonessential amino acid. In some embodiments, the amount of the nonessential amino acid is at least approximately 5 μM. The amount of the nonessential amino acid may be 5-600 μM. The nonessential amino acid may be selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, and tyrosine.
  • In certain embodiments, the liquid composition comprises 0-4 μM of cysteine and/or cystine. For example, the liquid composition may comprise 0-1 μM of cysteine and/or cystine.
  • In certain embodiments, the liquid composition has a pH of approximately 5.0-6.5. The liquid composition may have a pH of approximately 5.5-6.0. The liquid composition may have a pH of approximately 6.0. The liquid composition may have a pH of approximately 5.5. The liquid composition may have a pH of approximately 5.0.
  • In certain embodiments, the liquid composition has an osmolarity of 300-500. For example, the liquid composition may have an osmolarity of 300-400. The liquid composition may have an osmolarity of 400-500. The liquid composition may have an osmolarity of 300-350.
  • For the liquid composition, in certain embodiments, the aqueous solution is sterile. In another embodiment, the aqueous solution is nonsterile. In some embodiments, the liquid composition is a dialysis solution.
  • In some aspects, the present disclosure provides, a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the dry composition optionally comprises glutamate, wherein the dry composition comprises 0-5 μM of cysteine and/or cystine.
  • In some aspects, the present disclosure provides a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the dry composition optionally comprises glutamate, wherein the dry composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, and wherein the dry composition comprises 0-5 μM of cysteine and/or cystine.
  • In some embodiments, the carbohydrate is selected from a group consisting of dextrose, glucose, sucrose, fructose, and lactose. The carbohydrate may be dextrose.
  • In some aspects, the present disclosure provides a dry composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (c) at least one essential amino acid, wherein the dry composition comprises at least 2 μg of essential amino acid, and wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • For the dry composition, in certain embodiments, the amount of dextrose is at least approximately 10 g. The amount of dextrose may be 10-50 g, such as approximately 50 g. In another embodiment, the amount of dextrose is at least approximately 0.5 g. The amount of dextrose may be 0.5-2.5 g, such as approximately 2 g.
  • For the dry composition, in certain embodiments, the essential amino acid is selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine. In some embodiments, at least one essential amino acid is methionine. In some embodiments, at least one essential amino acid is arginine. In some embodiments, the dry composition comprises at least two essential amino acids. The dry composition may comprise at least four essential amino acids. The dry composition may comprise at least five essential amino acids. The dry composition may comprise at least six essential amino acids. The dry composition may comprise at least seven essential amino acids. The dry composition may comprise at least eight essential amino acids. The dry composition may comprise at least nine essential amino acids. The dry composition may comprise at least ten essential amino acids. In some embodiments, the dry composition comprises at least 2 μg of the essential amino acid.
  • The dry composition may comprise at least one vitamin. The vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline. In some embodiments, the amount of the vitamin is about 0.01-1500 μm. For example, the vitamin is vitamin C. The dry composition may comprise at least 1000 μg of vitamin C. In some embodiments, the dry composition comprises at least 2500 μg of vitamin C. In some embodiments, the dry composition comprises at least 0.01 μg of said vitamin B. The dry composition may comprise at least 1 μg of vitamin B.
  • In some embodiments, the amount of said glutamate is approximately 0.1 mg to 50 mg. The liquid composition may be free of nonessential amino acids other than glutamate.
  • The dry composition may further comprise an essential fatty acid, such as an omega-3 fatty acid.
  • The dry composition may further comprise at least one mineral. The mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc. In some embodiments, the dry composition comprises at least 0.3 μg of mineral. For example, the dry composition may comprise at least 1 μg of mineral.
  • In certain embodiments, the dry composition is stored in a container. The container may be a dialysis bag. The dialysis bag may be intended for single-use. The dry composition may be a dialysis mixture. In some embodiments, the dry composition further comprises an aqueous solution in the container, thereby producing a mixture. The container may comprise approximately 2000 mL of aqueous solution. The container may comprise approximately 2500 mL of aqueous solution. The mixture may have a pH of 4.0-6.5. The mixture may have an osmolarity of 300-720 mOsmol/L.
  • In certain embodiments, the amount of at least one ion is at least 100 μmol.
  • In certain embodiments, the dry composition further comprises at least one nonessential amino acid. The amount of nonessential amino acid may be at least 1 μg. In some embodiments, the amount of nonessential amino acid is 1-100 μg.
  • The dry composition may comprise 0-5 mg of cysteine and/or cystine. In some embodiments, the dry composition comprises 0-1 mg of cysteine and/or cystine.
  • In some aspects, the present disclosure provides a concentrated composition comprising a dry composition described herein, and an initial aqueous solution, wherein the dry composition is dissolved in the initial aqueous solution.
  • In some aspects, the present disclosure provides a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least one nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline, wherein each nonessential amino acid is 0-1% wt/wt of protein in the food composition, wherein the food composition comprises 0-1% cysteine and/or cystine wt/wt of protein in the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • For the food composition, in certain embodiments, the amount of nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline is 0-0.1% wt/wt of protein in the food composition. In some aspects, the food composition comprises at least two nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least three nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least four nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least five nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least six nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. In some embodiments, the nonessential amino acid is glycine. In some embodiments, the nonessential amino acid is proline.
  • In certain embodiments, the amount of cysteine is 0-0.1% wt/wt of protein in the food composition.
  • In some aspects, the present disclosure provides a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least six nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine, wherein each nonessential amino acid is 0-1% wt/wt of protein in the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • For the food composition, in certain embodiments, the amount of each nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine is 0-50%. In some embodiments, the food composition comprises at least seven nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least eight nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least nine of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least ten nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. In some embodiments, at least six nonessential amino acids are selected from the group consisting of glycine, proline, glutamine, serine, tyrosine, and cysteine. In an exemplary embodiment, at least one nonessential acid is cysteine. The amount of cysteine and/or cystine may be 0-0.04% wt/wt of protein in the food composition.
  • In some aspects, the present disclosure provides a food composition comprising: (a) at least one essential amino acid; and (b) glutamate, wherein the food composition is substantially free of nonessential amino acids other than glutamate.
  • In any of the food compositions disclosed herein, the amount of the cysteine devoid protein source is at least 1 wt/wt of the food composition. The amount of the cysteine devoid protein source may be 1-5% of the food composition. In an exemplary embodiment, the cysteine devoid protein source is gelatin.
  • In any of the food compositions disclosed herein, further comprising glutamate. In some embodiments, the food composition comprises approximately 0.1 mg to 50 mg of glutamate.
  • In any of the food compositions disclosed herein, the essential amino acid is selected from a group consisting of arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine. In an exemplary embodiment, the essential amino acid is methionine. The essential amino acid may be at least 1.0% wt/wt of the food composition. For example, the amount of essential amino acid may be 2.0-75% wt/wt of the food composition. In some embodiments, the food composition comprises at least two essential amino acids. The food composition may comprise at least three essential amino acids. The food composition may comprise at least four essential amino acids. The food composition may comprise at least five essential amino acids. The food composition may comprise at least six essential amino acids. The food composition may comprise at least seven essential amino acids. The food composition may comprise at least eight essential amino acids.
  • In certain embodiments, any food composition described herein further comprises a pharmaceutically active agent. The pharmaceutically active agent may be an anti-cancer therapeutic.
  • In any of the food compositions disclosed herein, the food composition is packaged as a solid food, semi-solid food, or beverage. The food composition is packaged as a food product selected from a group consisting of a snack bar, cereal product, bakery product, and dairy product. For example, the food composition may be baby formula. The food composition may be a soft drink. The food composition may be a nutritional shake. The food composition is a granola bar.
  • In some embodiments, the food composition comprises 10-20% w/w of total amino acids. The food composition may comprise approximately 17% w/w of total amino acids.
  • In some aspects, the present disclosure provides a facility comprising a plurality of the liquid compositions described herein. The facility may be a pharmaceutical manufacturing site. The facility may be a distribution center. The facility may be a hospital or clinic.
  • In some aspects, the present disclosure provides a plurality of the dry compositions described herein. The facility may be a pharmaceutical manufacturing site. The facility may be a distribution center. The facility may be a hospital or clinic.
  • In some aspects, the present disclosure provides a plurality of the food composition described herein. The facility may be a food manufacturing site. The facility may be a distribution center. The facility may be a commercial kitchen, restaurant or eating facility.
  • In some aspects, the present disclosure provides a kit comprising: (a) a pre-packaged liquid composition described herein; and (b) instructions for using the pre-packaged liquid composition. In some embodiments, the kit provides complete treatments for at least one week. For example, the kit may provide complete treatments for at least four weeks.
  • In some aspects, the present disclosure provides a kit comprising: (a) a pre-packaged dry composition or concentrated composition described herein; and (b) instructions for using the pre-packaged dry composition or concentrated composition. In some embodiments, the kit provides complete treatments for at least one week. For example, the kit provides complete treatments for at least four weeks.
  • In some aspects, the present disclosure provides a kit comprising: (a) a pre-packaged food composition described herein; and (b) instructions for using the pre-packaged food composition. In some embodiments, the kit provides complete meals for at least one day. For example, the kit may provide complete meals for at least five days. In some embodiments, the kit further comprises a pre-packaged liquid composition described herein. The kit may further comprise instructions for using the pre-packaged liquid composition. In another embodiment, the kit further comprises a pre-packaged dry composition described herein. The kit further comprises instructions for using the pre-packaged dry composition.
  • In some aspects, the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • In some aspects, the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • In some aspects, the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • In any of the methods for treating cancer, the cancer may be selected from the group consisting of liver cancer, breast cancer, lung cancer, leukemia, prostate, and colon cancer. The subject may be human. The subject may be an animal. In some embodiments, the subject has a genetic predisposition for cancer. The subject may have elevated cysteine biosynthetic gene expression levels.
  • In certain embodiments, treating cancer is characterized by a reduction in tumor size. The tumor may be reduced by at least 30%. The tumor may be reduced by at least 50%. The tumor may be reduced by at least 80%.
  • In some embodiments, treating cancer is characterized by decreased levels of plasma cysteine. The levels of plasma cysteine may be reduced by at least 30%. The levels of plasma cysteine may be reduced by at least 50%. The levels of plasma cysteine may be reduced by at least 80%. The levels of plasma cysteine may be reduced by at least 90%. In an exemplary embodiment, the levels of plasma cysteine are reduced by at least 95%.
  • In some aspects, the present disclosure provides a method for reducing tumor size in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • In some aspects, the present disclosure provides a method for reducing tumor size in a subject in need thereof, the method comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • In some aspects, the present disclosure provides a method for reducing tumor size in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • In any of the methods for reducing tumor size, the subject may be human. The subject may be an animal. In some embodiments, the subject has a genetic predisposition for cancer. The subject may have elevated cysteine biosynthetic gene expression levels.
  • In certain embodiments, reducing tumor size is characterized by a reduction in tumor size. The tumor may be reduced by at least 30%. The tumor may be reduced by at least 50%. The tumor may be reduced by at least 80%.
  • In some aspects, the present disclosure provides a method for treating obesity, the method comprising administering to the subject any food composition described herein.
  • In any of the methods for treating obesity, the subject may be human. The subject may be an animal. In some embodiments, the subject has a genetic predisposition for obesity.
  • In some embodiments, treating obesity is characterized by the subject losing at least 10% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 20% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 30% of the subject's initial body weight.
  • In some aspects, the present disclosure provides a method for treating renal disease, the method comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein.
  • In any of the methods for treating renal disease, the subject may be human. The subject may be an animal. In an exemplary embodiment, the subject is diagnosed with chronic kidney disease.
  • In any of the methods comprising administering to the subject a liquid composition, a concentrated composition or a dry composition described herein, the methods further comprise utilizing a membrane. In certain embodiments, the membrane comprises modified cellulose diacetate or polysulfone. In an exemplary embodiment, the membrane is a low-flux membrane. The low-flux membrane may permit small molecule exchange. In some embodiments, the low-flux membrane comprises a plurality of pores, and the plurality of pores comprises a first end and a second end. The plurality of pores may have an average pore diameter of approximately 1.8 nm.
  • In certain embodiments, the low-flux membrane permits approximately 50% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. The low-flux membrane may permit approximately 75% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. The low-flux membrane may permit approximately 90% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. The low-flux membrane may permit approximately 95% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end.
  • In certain embodiments, approximately 20% of a molecule of at least 1000 Daltons passes through the plurality of pores, from the first end to the second end. For example, approximately 10% of a molecule of at least 1000 Daltons may pass from the first side to the second side. Approximately 1% of a molecule of at least 1000 Daltons may pass from the first side to the second side.
  • In any of the methods comprising administering the liquid composition, concentrated composition, or dry composition, in certain embodiments, the composition is administered to the subject at least once a week. The composition may be administered to the subject at least three times a week. The composition may be administered to the subject at least five times a week. In certain embodiments, the liquid composition, concentrated composition, or dry composition is administered to the subject over the course of at least one week. The composition may be administered to the subject over the course of at least four weeks. The composition may be administered to the subject over the course of at least 2 months. In an exemplary embodiment, the composition is administered to the subject over the course of at least 3 months.
  • In any of the methods comprising administering any food composition described herein, in certain embodiments, the food composition is administered to the subject at least once a week. The food composition may be administered to the subject at least three times a week. The food composition may be administered to the subject at least five times a week. The food composition may be administered to the subject at least twenty-one times a week.
  • In certain embodiments, the food composition is administered to the subject over the course of at least one week. The food composition may be administered to the subject over the course of at least four weeks. The food composition may be administered to the subject over the course of at least 2 months. The food composition may be administered to the subject over the course of at least 3 months.
  • In any of the methods comprising administering a liquid composition, concentrated composition or dry composition, and any food composition, the compositions may be administered to the subject sequentially. In another embodiment, the compositions may be administered to the subject simultaneously. In some embodiments, the compositions are administered to the subject at least once a week. For example, the compositions may be administered to the subject at least three times a week. In some embodiments, the compositions are administered to the subject over the course of at least one week. For example, the compositions may be administered to the subject over the course of at least four weeks.
  • In any of the methods described herein, in certain embodiments, the methods further comprise administering a pharmaceutically active agent. The pharmaceutically active agent may be an anti-cancer therapeutic. The pharmaceutically active agent may be provided in a subtherapeutic amount.
  • In any of the methods described herein, in certain embodiments, the methods further comprise administering a nutritional supplement. The nutritional supplement may comprise at least one amino acid selected from a group consisting of arginine, histidine, isoleucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine. In some embodiments, the nutritional supplement provides the subject with nutrients at levels in accordance with the Recommended Dietary Allowances (RDAs), Adequate Intakes (AIs), and/or Daily Value (DV).
  • In some aspects, the disclosure provides a method of treating cancer in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • In some aspects, the disclosure provides a method of depleting cysteine in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • In some aspects, the disclosure provides a method of treating cancer in a subject that has been placed on a dialysis procedure to reduce cysteine in the blood, comprising: placing the subject on a low-cysteine diet.
  • In some embodiments, the dialysis comprises administering a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein. In some embodiments, the low-cysteine diet is a food composition disclosed herein.
  • In some aspects, the disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein.
  • In some aspects, the disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein, and a dialysis machine configured to administer the liquid composition, the concentrated composition or the dry composition. In some embodiments, the dialysis machine comprises a pump, a pressure monitor, air trap, air detector, a clamp, and/or a filter.
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • A description of various aspects, features, embodiments, and examples is provided herein with reference to the accompanying drawings, which are briefly described below. The drawings may illustrate one or more aspect(s), feature(s), embodiment(s), and/or example(s) in whole or in part. The drawings are illustrative and are not necessarily drawn to scale.
  • The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
  • FIG. 1 depicts a metabolic cancer therapy in which a cancer patient is treated with dialysis and/or a nutritional diet in order to deplete targeted nutrient(s).
  • FIG. 2 shows an overview of the diets used in colon, breast, prostate, and pancreatic cancer studies.
  • FIG. 3 shows an overview of the diet compositions used in the studies of the present disclosure. The mice used for the study were normal and active (other than tumor-bearing.)
  • FIG. 4 depicts a graph showing the effects of a complete diet and MEAD202 on tumor volume (mm3) in colon cancer (HCT116). Mice with implanted HCT116 cells were on diet treatment on day 7. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 5A depicts a graph showing the effects of a complete diet (Ctrl) and treatment diets on tumor volume (mm3) over time (days after tumor implant) in triple negative breast cancer (MDA-MDB-231). Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P <0.05, ** for P<0.01, *** for P<0.001. FIG. 5B depicts a graph showing the effects of a complete diet (Ctrl), MEAD202 and MEAD203 on tumor volume (mm3) over time (days after tumor inoculation) in triple negative breast cancer (MDA-MDB-231). Mice with implanted MDA-MDB-231 cells were on diet treatment on day 7. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 6 depicts a graph showing the effects of a complete diet (Ctrl) and MEAD203 on tumor volume (mm3) over time (days after tumor inoculation) in prostate cancer (PC3). Mice with implanted PC-3 cells were on diet treatment on day 5. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 7 depicts a graph showing the effects of a complete diet (Ctrl) and treatment diets on tumor volume (mm3) over time (days after treatment) in pancreatic cancer (MIAPaCa-2). Mice with implanted MIAPaCa-2 cells were on diet treatment on day 0. The tumor sizes were monitored 2 times weekly. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 8 shows an overview of the dialysis compositions used in the studies of the present disclosure.
  • FIG. 9 depicts a graph showing the effects of a complete diet (Ctrl), MEAD203+dialysis, MEAD203 on tumor volume (mm3) over time (days after tumor inoculation) in glioblastoma (U87MG, single mouse study). Mice with implanted U87MG cells were on diet treatment on day 5. The tumor sizes were monitored daily. Each treatment group had 1 mouse. The dialysis fluid was MEAD101 (Ctrl) at day 7, 8, 9, 13, 14, 28, 37, and 40. The dialysis fluid was MEAD102 at day 23, 26, 27, 30, 31, 33, 34, 35, 36, 41, and 42. The dialysis fluid was MEAD103 at 43, 44, 47, 48, 49, 50, 51, 54, 55, 56, 57 and 58.
  • FIG. 10 depicts a graph showing the effects of a complete diet, diet only, dialysis only, and diet+dialysis on tumor volume (mm3) over time (days after treatment) in glioblastoma (U87MG). Mice with implanted U87MG cells were on diet treatment on day 1. The tumor sizes were monitored daily. Each group had 6 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 11A depicts a graph showing the effects of a complete diet (Ctrl), MEAD203, MEAD202, and natural chow on body weight (%) over time (days after tumor inoculation) in triple negative breast cancer (MDA-MB-231). Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. FIG. 11B depicts a graph showing the effects of a complete diet (Ctrl), and MEAD203 on body weight (%) over time (days after tumor inoculation) in prostate cancer (PC3). Mice with implanted PC-3 cells were on diet treatment on day 5. The tumor sizes were monitored 3 times weekly. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001
  • FIG. 12A depicts a graph showing the effects of natural chow, complete diet, MEAD202, MEAD203 on glucose (mg/dL) in triple negative breast cancer (MDA-MB-231). Glucose in plasma was measured with Nova Max Glucose Test Strips. Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. FIG. 12B depicts a graph showing the effects of complete diet and MEAD203 on glucose (mg/dL) in prostate cancer (PC3). Glucose in plasma was measured with Nova Max Glucose Test Strips. Mice with implanted PC-3 cells were on diet treatment on day 5. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 13A depicts a graph showing the effects of natural chow, complete diet, MEAD202, MEAD203 on β-Ketone (mmol/L) in triple negative breast cancer (MDA-MB-231). Beta-ketone body in plasma was measured with Nova Max Ketone Test Strips. Mice with implanted MDA-MDB-231 cells were on diet treatment on day 20. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. FIG. 13B depicts a graph showing the effects of complete diet and MEAD203 on β-Ketone (mmol/L) in prostate cancer. Beta-ketone body in plasma was measured with Nova Max Ketone Test Strips. Mice with implanted PC-3 cells were on diet treatment on day 5. The plasma samples were collected before treatment started and after treatment completed. Each group had 5 mice. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • FIG. 14A and FIG. 14B show exemplary dialysis systems, CAPD and CCPD respectively, that is configured to use administer the compositions described herein.
  • FIG. 15 shows and exemplary dialysis system for hemodialysis that is configured to use administer the compositions described herein.
  • FIG. 16 depicts the typical hemodialysis process.
  • DETAILED DESCRIPTION
  • Several aspects of the invention are described below with reference to example applications for illustration. It should be understood that numerous specific details, relationships, and methods are set forth to provide a full understanding of the invention. One having ordinary skill in the relevant art, however, will readily recognize that the invention can be practiced without one or more of the specific details or with other methods. Unless stated otherwise, the present invention is not limited by the illustrated ordering of acts or events, as some acts may occur in different orders and/or concurrently with other acts or events. Furthermore, not all illustrated acts or events are required to implement a methodology in accordance with the present invention. The concentration of various components in the disclosed compositions is exemplary and not meant to be limited to the recited concentration per se.
  • It will be understood that a word appearing herein in the singular encompasses its plural counterpart, and a word appearing herein in the plural encompasses its singular counterpart, unless implicitly or explicitly understood or stated otherwise. Further, it will be understood that for any given component described herein, any of the possible candidates or alternatives listed for that component, may generally be used individually or in any combination with one another, unless implicitly or explicitly understood or stated otherwise. Additionally, it will be understood that any list of such candidates or alternatives, is merely illustrative, not limiting, unless implicitly or explicitly understood or stated otherwise. Still further, it will be understood that any figure or number or amount presented herein is approximate, and that any numerical range includes the minimum number and the maximum number defining the range, whether the word “inclusive” or the like is employed or not, unless implicitly or explicitly understood or stated otherwise. Generally, the term “approximately” or “about” or the symbol “—” in reference to a figure or number or amount includes numbers that fall within a range of ±5% of same, unless implicitly or explicitly understood or stated otherwise. Yet further, it will be understood that any heading employed is by way of convenience, not by way of limitation. Additionally, it will be understood that any permissive, open, or open-ended language encompasses any relatively permissive to restrictive language, less open to closed language, or less open-ended to closed-ended language, respectively, unless implicitly or explicitly understood or stated otherwise. Merely by way of example, the word “comprising” may encompass “comprising”, “consisting essentially of”, and/or “consisting of” type language.
  • As used herein, unless otherwise indicated, some inventive embodiments herein contemplate numerical ranges. A variety of aspects of this invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range as if explicitly written out. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range. When ranges are present, the ranges include the range endpoints.
  • Definitions
  • The terms “administer”, “administered”, “administers” and “administering” are defined as the providing a composition to a subject via a route known in the art, including but not limited to intravenous, intraarterial, oral, parenteral, buccal, topical, transdermal, rectal, intramuscular, subcutaneous, intraosseous, transmucosal, or intraperitoneal routes of administration. In certain embodiments of the subject application, oral routes of administering a composition may be preferred. In certain embodiments, administering a composition through dialysis may be preferred.
  • As used herein, “agent” or “biologically active agent” refers to a biological, pharmaceutical, or chemical compound or other moiety. Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, a peptide nucleic acid (PNA), an oligonucleotide (including e.g., aptamer and polynucleotides), an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, a branched chain amino acid in free amino acid form or metabolite thereof, or a chemotherapeutic compound. As used herein, the term protein includes an amino acid mixture comprising one or more amino acids. Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures. In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • As used herein, the terms “co-administration”, “administered in combination with” and their grammatical equivalents are meant to encompass administration of the invention composition and additional therapeutic agent to a single subject. Co-administration can encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the subject at the same time. Co-administration can encompass treatment regimens in which the composition and additional therapeutic agent are administered by the same or different route of administration or at the same or different times. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present. Co-administration can include simultaneous administration of the agents in separate compositions, administration at different times in separate compositions, and/or administration in a single composition comprising each of the agents to be co-administered.
  • The terms “determining”, “measuring”, “evaluating”, “assessing,” “assaying,” “interrogating,” and “analyzing” are used interchangeably herein to refer to any form of measurement, and include determining if an element is present or not. These terms include both quantitative and/or qualitative determinations. Assessing may be relative or absolute.
  • The term “effective amount” or “therapeutically effective amount” refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below. The therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or down regulation of activity of a target protein. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • The term “essential amino acid” or “indispensable amino acid” refers to an amino acid that cannot be synthesized de novo by the organism. Essential amino acids include arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine. Essential amino acids may include salts thereof.
  • The term “free”, as used herein, refers to compositions that have about 0% of a specific component.
  • The term “nonessential amino acid” or “dispensable amino acid” refers to an amino acid that can be synthesized de novo by the organism. Nonessential amino acids include alanine, asparagine, aspartate, cysteine (or cystine), glutamic acid, glutamine, glycine, proline, serine, and tyrosine. The terms “cysteine” and “cystine” are used interchangeably herein. Nonessential amino acids may also include salts thereof.
  • The term “prevention” refers to providing treatment prior to the onset of a condition. If treatment is commenced in subjects with a condition, such treatment is expected to prevent, or to prevent the progression of, the medical sequelae of the condition.
  • As used herein, the term “subject” or “individual” includes mammals. Non-limiting examples of mammals include humans and animals, such as mice, including transgenic and non-transgenic mice. The methods described herein can be useful in both human therapeutics, pre-clinical, and veterinary applications. In some embodiments, the subject is an animal, and in some embodiments, the subject is human. Other mammals include, and are not limited to, apes, chimpanzees, orangutans, monkeys; domesticated animals (pets) such as dogs, cats, guinea pigs, hamsters, mice, rats, rabbits, and ferrets; domesticated farm animals such as cows, buffalo, bison, horses, donkey, swine, sheep, and goats; or exotic animals typically found in zoos, such as bear, lions, tigers, panthers, elephants, hippopotamus, rhinoceros, giraffes, antelopes, sloth, gazelles, zebras, wildebeests, prairie dogs, koala bears, kangaroo, pandas, giant pandas, hyena, seals, sea lions, and elephant seals.
  • The term “substantially free”, as used herein, refers to compositions that have less than about 10%, less than about 5%, less than about 1%, less than about 0.5%, less than 0.1% or even less of a specified component.
  • A “subtherapeutic amount” or “sub-therapeutic amount” of an agent, an activator or a therapy is an amount less than the effective amount for that agent, activator or therapy, but when combined with an effective or sub-therapeutic amount of another agent or therapy can produce a desired result, due to, for example, synergy in the resulting efficacious effects, and/or reduced side effects. A sub-therapeutic amount of the agent or component can be such that it is an amount below which would be considered therapeutic. For example, FDA guidelines can suggest a specified level of dosing to treat a particular condition, and a sub-therapeutic amount would be any level that is below the FDA suggested dosing level. The sub-therapeutic amount can be about 1, 5, 10, 15, 20, 25, 30, 35, 50, 75, 90, or 95% less than the amount that is considered to be a therapeutic amount. The therapeutic amount can be assessed for individual subjects, or for groups of subjects. The group of subjects can be all potential subjects, or subjects having a particular characteristic such as age, weight, race, gender, or physical activity level.
  • Compositions
  • As used herein “composition” refers to a dry composition, a concentrated composition, a liquid composition, and/or a food composition. The present disclosure provides for compositions, such as dry compositions, concentrated compositions, liquid compositions, and food compositions, effective for treating cancer, other metabolic diseases, including obesity, and renal disease. The compositions can be depleted or reduced with respect to one or more amino acids or other nutrients and/or supplemented with respect to one or more other amino acids or nutrients. The depleted or reduced nutrients can be certain nutrients acids that are demanded at a higher degree by cancerous cells as compared to non-cancerous cells. For example, cancerous cells may have a higher nutritional or metabolic requirement for certain non-essential amino acids as compared to non-cancerous cells and so a composition may formulated to be depleted or reduced with respect to those non-essential amino acids while maintaining the normal or supplemented amounts of other nutrients. The compositions may be a composition for dialysis or for a nutritional diet. As used herein, a dry composition, a concentrated composition, or a liquid composition may also be referred as a “dialysate composition,” “dialysate,” a “dialysis solution,” or a “dialysis composition. As used herein, a food composition may also be referred to as a “nutritional diet” or a “diet.”
  • Liquid Composition
  • The present disclosure provides dialysis compositions. The dialysis composition may be in the form of a liquid composition. In some aspects, the present disclosure provides a liquid composition comprising glutamate. In some embodiments, the liquid composition comprises glutamate and at least one essential amino acid. The present disclosure also provides a dialysis composition comprising at least one vitamin. In some embodiments, the liquid composition comprises at least one vitamin and at least one essential amino acid. In some embodiments, the liquid composition is substantially free of cysteine and/or cystine.
  • In some aspects, the present disclosure provides, a liquid composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one vitamin; (d) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (e) at least one essential amino acid; and (f) an aqueous solution, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L. In some aspects, the present disclosure provides, a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some aspects, the present disclosure provides, a liquid composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition is substantially free of nonessential amino acids other than glutamate, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L. In some aspects, the present disclosure provides a liquid composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the liquid composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, wherein the liquid composition comprises 0-5 μM of cysteine and/or cystine, wherein said liquid composition has a pH of 4.0-6.5, and wherein said liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some aspects, the present disclosure provides a liquid composition comprising: (a) carbohydrate; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (c) at least one essential amino acid; and an aqueous solution, wherein the liquid composition comprises 0-50 μM of cysteine and/or cystine, wherein liquid composition has a pH of 4.0-6.5, and wherein the liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some aspects, the present disclosure provides a liquid composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (c) at least one essential amino acid; and an aqueous solution, wherein the liquid composition comprises at least 10 μM of essential amino acid, wherein the liquid composition comprises 0-50 μM of cysteine and/or cystine, wherein liquid composition has a pH of 4.0-6.5, and wherein the liquid composition has an osmolarity of 300-720 mOsmol/L.
  • In some aspects, the present disclosure provides a liquid composition comprising: (a) carbohydrate; (b) at least one essential amino acid; and an aqueous solution, wherein the liquid composition comprises 0-50 μM of cysteine and/or cystine, and wherein the liquid composition is substantially free of nonessential amino acids.
  • In some embodiments, the carbohydrate is selected from a group consisting of dextrose, glucose, sucrose, fructose, and lactose. In an exemplary embodiment, the carbohydrate is dextrose. The liquid composition disclosed herein may comprise amount of dextrose that is 0.1-4.5% w/v. In certain embodiments, the amount of the dextrose is 1.0-4.5% w/v. The amount of the dextrose may be 1.5-4.25% w/v. In an exemplary embodiment, the amount of the dextrose is 1.7-4.0% w/v. In another embodiment, the amount of dextrose is 0.05-0.25% w/v. The amount of the dextrose may be 0.08-0.22% w/v. In an exemplary embodiment, the amount of the dextrose is 0.1-0.2% w/v. The amount of dextrose in the liquid composition may be at least, approximately, or no more than 0.05, 0.1, 0.5, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, or 5.0% w/v. The amount of dextrose in the liquid composition may depend on its intended application, such as use in peritoneal dialysis (PD) or use in hemodialysis (HD). For example, when used for peritoneal dialysis, the amount of dextrose in the liquid composition may be 1.0-4.5% w/v, such as 1.5-4.25% w/v or 1.7-4.0% w/v. When used for hemodialysis, the amount of dextrose in the liquid composition may be 0.05-0.50% w/v, such as 0.08-0.25% w/v or 0.1-0.25% w/v.
  • In certain embodiments, the liquid composition comprises at least one essential amino acid selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine. In an exemplary embodiment, at least one of the essential amino acids is methionine. In an exemplary embodiment, at least one of the essential amino acids is arginine. In certain embodiments, the amount of at least one of the essential amino acid is at least 10 μM. The amount of at least one of the essential amino acid may be 10-300 μM. The amount of at least one of the essential amino acid may be 20-250 μM. The amount of at least one of the essential amino acid may be at least, approximately, or no more than 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, or 300 μM.
  • In certain embodiments, the liquid composition comprises at least two essential amino acids. The liquid composition may comprise at least three essential amino acids. The liquid composition may comprise at least four essential amino acids. The liquid composition may comprise at least five essential amino acids. The liquid composition may comprise at least six essential amino acids. The liquid composition may comprise at least seven essential amino acids. The liquid composition may comprise at least eight essential amino acids. The liquid composition may comprise at least nine essential amino acids. The liquid composition may comprise at least ten essential amino acids.
  • In certain embodiments, the liquid composition comprises at least one vitamin. The vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline. In some embodiments, the amount of vitamin is at least, approximately, or no more than about 0.00001, 0.0001, 0.001, 0.01, 0.1, 1, 25, 50, 75, 100, 125, 150, 175 or 200 μM. The amount of vitamin may be approximately about 0.000010 to 200 μM. In some embodiments, the liquid composition comprises approximately at least 0.001, 100, or 1000 μM of at least one vitamin. In an exemplary embodiment, the vitamin is vitamin C. In one embodiment, the liquid composition comprises at least approximately 1 μM of the vitamin C. The liquid composition may comprise at least approximately 7 μM of the vitamin C. The liquid composition may comprise at least approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 μM of vitamin C. In an exemplary embodiment, the vitamin is vitamin B. In some embodiments, the liquid composition comprises 0.000005 to 1500 μM of said vitamin B, such as 0.00005, 0.0005, 0.005, 0.05, 0.5, 1, 10, 100, 1000 or 1500 μM. Vitamin B may include biotin, vitamin B1, vitamin B2, vitamin B3, vitamin B5, vitamin B6, vitamin B9, and/or vitamin B12.
  • In certain embodiments, the liquid composition comprises glutamate. The amount of glutamate may be at least, approximately, or no more than 1, 5, 20, 50, 100, 200, 300, 400, 500, 750, or 1000 μM. The amount of glutamate may be approximately 2-500, 20-400, 75-300, or 100-200 μM. In some embodiments, the amount of glutamate increases glutamate in the blood by at least, approximately or no more than 25, 50, 75, 100, 125, 150, 175, 200, or 300 μM. In certain embodiments, the amount of glutamate increases glutamate in the blood by at least 150 μM.
  • In certain embodiments, the liquid composition further comprises an essential fatty acid, such as omega-6 and omega-3 fatty acids. In one embodiment, the essential fatty acid is an omega-3 fatty acid. The amount of essential fatty acid may be approximately 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1500, or 2000 mg per liter. In some embodiments, the amount of essential amino acids is 200-350 mg per liter.
  • In certain embodiments, the liquid composition further comprises at least one mineral. The mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc. In an embodiment, the liquid composition comprises at least 5 nM of the mineral. The amount of at least one of the mineral may be 5 nM-5 μM. In an exemplary embodiment, the liquid composition comprises 1 μM or more.
  • The liquid composition described herein comprises at least one ion. In certain embodiments, the amount of at least one ion is at least approximately 0.1 mEq/L. The amount of at least one ion may be 0.1-150 mEq/L. The amount of at least one ion may be at least approximately 0.1, 0.5, 1, 5, 10, 15, 20, 50, 75, 100, 125, or 150 mEq/L.
  • In some embodiments, the liquid composition is substantially free of at least one nonessential amino acid selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine. The liquid composition may be substantially free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids. The liquid composition may be free of at least one nonessential amino acid. The liquid composition may be free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids. The liquid composition may be free of nonessential amino acids other than glutamate. Alternatively, in certain embodiments, the liquid composition further comprises at least one nonessential amino acid. The nonessential amino acid can be selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, and tyrosine. In an embodiment, the liquid composition comprises at least two nonessential amino acids. The liquid composition may comprise at least three nonessential amino acids. The liquid composition may comprise at least four nonessential amino acids. The liquid composition may comprise at least five nonessential amino acids. The liquid composition may comprise at least six nonessential amino acids. The liquid composition may comprise at least seven nonessential amino acids. The liquid composition may comprise at least eight nonessential amino acids. The liquid composition may comprise at least nine nonessential amino acids. In one embodiment, the amount of the nonessential amino acid is at least 5 approximately μM, such as 5, 10, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or 700 μM. The amount of the nonessential amino acid may be 5-600 μM.
  • The liquid composition may comprise less than approximately 5 μM of cysteine and/or cystine. For example, the amount of cysteine and/or cystine may be 0, 0.1, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, or 5 μM. In certain embodiments, the liquid composition comprises 0-4 μM of cysteine and/or cystine. The liquid composition may comprise 0-2.5 μM of cysteine and/or cystine, such as 0-1 μM of cysteine and/or cystine. In an exemplary embodiment, the liquid composition is substantially free of cysteine and/or cystine. The liquid composition may be free of cysteine and/or cystine.
  • In certain embodiments, the liquid composition has a pH of 5.0-6.5. The liquid composition may have a pH of 5.5-6.0. For example, the liquid composition may have a pH of approximately 6.0. The liquid composition may have a pH of approximately 5.5. The liquid composition may have a pH of approximately 5.0.
  • In certain embodiments, the liquid composition has an osmolarity of 300-500. The liquid composition may have an osmolarity of 300-400. In one embodiment, the liquid composition has an osmolarity of 300-350. The liquid composition may have an osmolarity of 400-500.
  • The liquid composition contains an aqueous solution that may be a sterile solution or a nonsterile solution. In some embodiments, the aqueous solution is sterile. In other embodiments, the aqueous solution is nonsterile. The liquid composition may be stored in a container that is ready to be used by the subject. The container may comprise a sufficient amount of liquid composition for use in any of the methods described herein. For example, the container may hold approximately 1, 2, or 2.5 liter of liquid composition. The liquid composition may be available as a ready-for-use dialysis solution.
  • Dry Composition/Concentrated Composition
  • The present disclosure provides dialysis compositions. The dialysis composition may be in the form of a dry or concentrated composition. In some aspects, the present disclosure provides a dry or concentrated composition comprising glutamate. In some embodiments, the dry or concentrated composition comprises glutamate and at least one essential amino acid. The present disclosure also provides a dialysis composition comprising at least one vitamin. In some embodiments, the dry or concentrated composition comprises at least one vitamin and at least one essential amino acid. In some embodiments, the dry or concentrated composition is substantially free of cysteine and/or cystine.
  • In some aspects, the present disclosure provides, a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the dry composition optionally comprises glutamate, wherein the dry composition comprises 0-5 μM of cysteine and/or cystine. In some aspects, the present disclosure provides, a dry composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one vitamin; (d) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (e) at least one essential amino acid, wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • In some aspects, the present disclosure provides a dry composition comprising: (a) carbohydrate; (b) at least one vitamin; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; (d) at least one essential amino acid; and (e) an aqueous solution, wherein the liquid composition optionally comprises glutamate, wherein the dry composition is substantially free of nonessential amino acids selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine, and wherein the dry composition comprises 0-5 μM of cysteine and/or cystine. In some aspects, the present disclosure provides, a dry composition comprising: (a) carbohydrate; (b) glutamate; (c) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (d) at least one essential amino acid, wherein the dry composition is substantially free of nonessential amino acids other than glutamate, and wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • In some aspects, the present disclosure provides a dry composition comprising: (a) dextrose; (b) at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate; and (c) at least one essential amino acid, wherein the dry composition comprises at least 2 μg essential amino acid, and wherein the dry composition comprises 0-10 mg of cysteine and/or cystine.
  • In some aspects, the present disclosure provides a dry composition comprising: (a) carbohydrate; (b) at least one essential amino acid, wherein the dry composition comprises 0-10 μg of cysteine and/or cystine.
  • The dry composition described herein may be advantageous since it may minimize storage space, weight, and transport of water and may also increase the stability and thereby also the shelf-life of the dialysis components. The dry composition may be stable for long term storage and transportation. The dry composition may be available in a container, in which an aqueous solution may be added to the container to reconstitute the contents therein. The container may have the capacity to hold approximately 1, 2, 2.5, 4 or 10 liter of aqueous solution. Aqueous solution may be added to the container prior to the administration of the composition to the subject for dialysis.
  • In some embodiments, the carbohydrate is selected from a group consisting of dextrose, glucose, sucrose, fructose, and lactose. In an exemplary embodiment, the carbohydrate is dextrose. The amount of dextrose in the dry composition may range from 0.1 g to 75 g. In certain embodiments, the amount of dextrose is approximately 5-75 g. The amount of the dextrose may be approximately 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, or 75 g. The amount of the dextrose may be 10-50 g. In one embodiment, the amount of dextrose is approximately 10 g. In one embodiment, the amount of dextrose is approximately 50 g. In another embodiment, the amount of dextrose is 0.1 g-5 g. The amount of the dextrose may be approximately 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5 or 5.0 g. The amount of the dextrose may be 0.5-2.5 g. In one embodiment, the amount of dextrose is approximately 0.5 g. In one embodiment, the amount of dextrose is approximately 2 g. In certain embodiments, the amount of dextrose is approximately 10-99.5% of the total composition. For example, the amount of dextrose may be 10, 20, 30, 40, 50, 60, 70, 75, 80, 85, 90, 95, or 99.5% of the total composition. In an exemplary embodiment, the amount of dextrose is 70-95% of the total composition.
  • In certain embodiments, the dry composition comprises at least one essential amino acid selected from a group consisting of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, and arginine. In an exemplary embodiment, at least one of the essential amino acid is methionine. In an exemplary embodiment, at least one of the essential amino acid is arginine. In certain embodiments, the amount of each essential amino acid is at least 2 μg. The amount of each essential amino acid may be at least, approximately, or no more than 1, 2, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 300, 500, 1000, or 1500 μg. The total amount of the essential amino acids may be at least, approximately, or no more than 100, 150, 200, 250, 300, 500, 1000, 2000, 3000, 5000, 6000, 7000, 7500 or 8000 μg.
  • In an embodiment, the dry composition comprises at least two essential amino acids. The dry composition may comprise at least three essential amino acids. The dry composition may comprise at least four essential amino acids. The dry composition may comprise at least five essential amino acids. The dry composition may comprise at least six essential amino acids. The dry composition may comprise at least seven essential amino acids. The dry composition may comprise at least eight essential amino acids. The dry composition may comprise at least nine essential amino acids. The dry composition may comprise at least ten essential amino acids.
  • In certain embodiments, the dry composition comprises at least one vitamin. The vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline. In some embodiments, the amount of vitamin is at least, approximately, or no more than 0.01, 0.05, 0.1, 0.5, 1, 10, 50, 100, 500, 750, 1000, 1250, or 1500 μg. The amount of vitamin may be approximately 0.01-1500 μm. In an exemplary embodiment, the vitamin is vitamin C. The dry composition may comprise at least approximately 1000 μg of vitamin C, such as approximately 1000, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, or 3000 μg of vitamin C. In an embodiment, the dry composition comprises at least approximately 2500 μg of vitamin C. The dry composition may comprise at least, approximately, or no more than 0.01 μg of vitamin B, such as approximately 0.01, 0.05, 0.1, 0.5, 1, 1.5, 5, 10, 50, or 100 μg of vitamin B. Vitamin B may include biotin, vitamin B1, vitamin B2, vitamin B3, vitamin B5, vitamin B6, vitamin B9, and/or vitamin B12.
  • In certain embodiments, the dry composition comprises at least, approximately, or no more than 0.1 mg of glutamate, such as approximately 0.1, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 mg. The amount of glutamate may be 0.1-50 mg, 5-25 mg, or 25-45 mg.
  • In certain embodiments, the dry composition further comprises an essential fatty acid, such as omega-6 and omega-3 fatty acids. In one embodiment, the essential fatty acid is an omega-3 fatty acid. In one embodiment, the essential fatty acid is an omega-3 fatty acid. The amount of essential fatty acid may be approximately 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 mg per liter. In some embodiments, the amount of essential amino acids is 1200-1600 mg per liter.
  • In certain embodiments, the dry composition further comprises at least one mineral. The mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc. In an embodiment, the dry composition comprises at least 0.3 μg of the mineral. For example, the dry composition comprises approximately 0.1, 1, 10, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1250, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000 or 7500 μg. In an exemplary embodiment, the dry composition comprises approximately 400 μg. In another embodiment, the dry composition comprises approximately 800 μg.
  • The dry composition described herein comprises at least one ion. The ion may be, but are not limited to, sodium, calcium, magnesium, chloride, lactate or combinations thereof. In certain embodiments, the amount of at least one of the ion is at least approximately 0.1 μg. In one embodiment, the amount of at least one of the ion is at least approximately 0.5 μg. The amount of the ion may be at least approximately 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 μg. In certain embodiments, the amount of the ion is at least approximately 50 μmol. In one embodiment, the amount of ion is at least approximately 100 μmol. The amount of ion may be at least approximately 200 μmol. The amount of ion may be at least approximately 250 μmol. The amount of the ion may be 50-300 μmol, such as 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, or 300 μmol.
  • In some embodiments, the dry composition is substantially free of at least one nonessential amino acid selected from alanine, asparagine, aspartate, cysteine, glycine, proline, glutamine, serine, and tyrosine. The dry composition may be substantially free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids. The dry composition may be free of at least one nonessential amino acid. The dry composition may be free of at least 2, 3, 4, 5, 6, 7, 8, or 9 nonessential amino acids. The dry composition may be free of nonessential amino acids other than glutamate. Alternatively, in certain embodiments, the dry composition further comprises at least one nonessential amino acid. The nonessential amino acid can be selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, and tyrosine. In an embodiment, the dry composition comprises at least two nonessential amino acids. The dry composition may comprise at least three nonessential amino acids. The dry composition may comprise at least four nonessential amino acids. The dry composition may comprise at least five nonessential amino acids. The dry composition may comprise at least six nonessential amino acids. The dry composition may comprise at least seven nonessential amino acids. The dry composition may comprise at least eight nonessential amino acids. The dry composition may comprise at least nine nonessential amino acids. In one embodiment, the amount of the nonessential amino acid is at least 1 mg. The amount of the nonessential amino acid may be 1-100 mg. In some embodiments, the amount of nonessential amino acid is 1-300 mg, such as 1, 50, 100, 150, 200, 250, or 300 mg.
  • The present disclosure provides a dry composition comprising less than 10 mg of cysteine and/or cystine. For example, the dry composition may comprise approximately 0, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 or 10 mg of cysteine and/or cystine. In certain embodiments, the dry composition comprises 0-10 mg of cysteine and/or cystine. In one embodiment, the dry composition comprises 0-2.5 mg of cysteine and/or cystine. In an exemplary embodiment, the dry composition is substantially free of cysteine and/or cystine. The dry composition may be free of cysteine and/or cystine.
  • In an exemplary embodiment, the liquid composition or dry composition comprises methionine. The molar ratio of dextrose to methionine of the liquid composition or dry composition may be greater or less than approximately 1, 50, 100, 250, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 10,500, 20,000, 30,000, 40,000, 50,500, 60,000, 70,000, 80,000, 90,000, or 100,000. In an exemplary embodiment, the molar ratio of dextrose to methionine of the liquid composition or dry composition is greater or less than approximately 250, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, or 10,500. In one embodiment, the molar ratio of dextrose to methionine of the compositions is approximately 250. In another embodiment, the molar ratio of dextrose to methionine of the compositions is approximately 10,500.
  • The molar ratio of cysteine to essential amino acids of the liquid composition or dry composition may be less than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 10, 15, 20, 50, or 100. In one embodiment, the molar ratio of cysteine to methionine is less than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • The molar ratio of methionine to amino acids of the liquid composition or dry composition may be greater than about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 10, 15, 20, 50, or 100. In some embodiments, the molar ratio of methionine to other amino acids of the liquid composition and dry composition may be greater than about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • In some embodiments, the molar ratio of methionine to alanine in the liquid composition or dry composition is greater than about 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to arginine is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to asparagine is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to aspartic acid is greater than about 2, 3, 4, 5, 10, 15, 20, 50, or 100. In some embodiments, the molar ratio of methionine to β-alanine is greater than about 1, 2, 3, 4, 5, 10, 15, 20, 50, or 100. In some embodiments, the molar ratio of methionine to cystine is greater than about 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to glutamic acid is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to glutamine is greater than about 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to glycine is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to histidine is greater than about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to isoleucine is greater than about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to leucine is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to phenylalanine is greater than about 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to proline is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to serine is greater than about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to threonine is greater than about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to tryptophan is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to tyrosine is greater than about 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5. In some embodiments, the molar ratio of methionine to valine is greater than about 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, or 5.
  • In certain embodiments, the molar ratio of methionine to minerals in the dry composition or liquid composition, such as iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium and zinc, of the liquid composition or dry composition is greater than about 1, 5, 7, 8, 9, 10, 40, 50, 75, 90, 100, 500, 1,000, 1,100, 1,500, 1,900, 2,000, 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000. The molar ratio of methionine to iron may be greater than about 9, 10, 40, 50, 75, 90, 100, 500, or 1,000. The molar ratio of methionine to copper may be greater than about 90, 100, 500, 1,000, 1,100, 1,500, 1,900, or 2,000. The molar ratio of methionine to chromium may be greater than about 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000. The molar ratio of methionine to fluoride may be greater than about 7, 8, 9, 10, 40, 50, 75, 90, 100, 500, or 1,000. The molar ratio of methionine to iodide may be greater than about 1,100, 1,500, 1,900, 2,000, 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000. The molar ratio of methionine to manganese may be greater than about 40, 50, 75, 90, 100, 500, or 1,000. The molar ratio of methionine to molybdenum may be greater than about 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000. The molar ratio of methionine to selenium may be greater than about 1,900, 2,000, 2,300, 2,500, 2,800, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000. The molar ratio of methionine to zinc may be greater than about 8, 9, 10, 40, 50, 75, 90, 100, 500, or 1,000.
  • In some embodiments, the dry composition, concentrated composition or liquid composition comprises one or more components, such as dextrose, ions, buffer, amino acids, vitamins and minerals, in terms of molar ratio (dextrose to component), as depicted in Table 1 below.
  • TABLE 1
    Dry Composition, Concentrated Composition and Liquid Composition Components
    Dextrose (/L) mmole
    Hemodialysis only (HD) 5.5-11.0
    peritoneal dialysis only (PD) 83-236
    Osmolarity (mOsmol/L) 300-718 
    pH 6.0 (4.0-6.5)  
    Molar Ratio Range Molar Ratio Range
    Ionic Concentration (/L) mmole (dextrose to ion) HD (dextrose to ion) PD
    Sodium 132 0.042-0.083 0.63-1.79
    Calcium 1.75 3.1-6.3 47.4-135 
    Potassium 2 2.75-5.5  41.5-118 
    Magnesium 0.25 22-44 332-944
    Chloride 96 0.057-0.11  0.86-2.46
    Molar Ratio Range Molar Ratio Range
    Buffer (L) mmole (dextrose to buffer) HD (dextrose to ion) PD
    HCO3 20-50 0.11-0.55 1.66-11.8
    Acetate 2.0-5.0 1.1-5.5 16.6-118 
    Lactate 40 0.14-0.28 2.1-5.9
    Molar Ratio Range Molar Ratio Range
    Amino acid (/L) micromole (dextrose to amino acid) HD (dextrose to ion) PD
    Alanine 347.5 16-32 239-679
    Arginine 60.25  91-183 1,378-3,917
    Asparagine 57.5  96-191 1,443-4,104
    Aspartic acid 8 688-1,375 10,375-29,500
    Beta-alanine 19.5 282-564  4,256-12,103
    Cystine 40.25 137-273 2,062-5,863
    Glutamic acid 72  76-153 1,153-3,278
    Glutamine 547.5 10-20 152-431
    Glycine 212.5 26-52 391-1,111
    Histidine 83.5  66-132 994-2,826
    Isoleucine 79.75  69-138 1,041-2,959
    Leucine 119 46-92 697-1,983
    Lysine 195 28-56 426-1,210
    Methionine 22.25 247-494  3,730-10,607
    Phenylalanine 55.25 100-199 1,502-4,271
    Proline 222.5 25-49 373-1,061
    Serine 109.75  50-100 756-2,150
    Threonine 137.25 40-80 605-1,719
    Tryptophan 66.5  83-165 1,248-3,549
    Tyrosine 63.75  86-173 1,302-3,702
    Valine 242.5 23-45 342-973
    Molar Ratio Range Molar Ratio Range
    Vitamin (/L) micromole (dextrose to vitamin) HD (dextrose to ion) PD
    Vitamin A 0.052 105,769-211,538 1,596,154-4,538,462
    Vitamin C 7.57 727-1,453 10,964-31,176
    Vitamin B6 1.48 3,716-7,432  56,081-159,459
    Vitamin E 0.90  6,111-12,222  92,222-262,222
    Folate (Vit B9) 15.18 362-725  5,468-15,547
    Vitamin D 0.0044 1,250,000-2,500,000 18,863,636-53,636,364
    Vitamin K 0.0038 1,447,368-2,894,737 21,842,105-62,105,263
    Thiamine 0.075  73,333-146,667 1,106,667-3,146,667
    Riboflavin 0.053 103,774-207,547 1,566,038-4,452,830
    Niacin 4.71 1,168-2,335 17,622-50,106
    Vitamin B12 0.000030 183,333,333-366,666,667 2,766,666,667-7,866,666,667
    Pantothenic acid 0.38 14,474-28,947 218,421-621,053
    Biotin 0.0021 2,619,048-5,238,095  39,523,810-112,380,952
    Choline 79.99  69-138 1,038-2,950
    105,769-211,538 1,596,154-4,538,462
    Mineral (/L)
    Iron 2.3279    2-4,725  35,654-101,379
    Copper 0.2360 23,305-46,610 351,695-1,000,000
    Chromium 0.0096 572,917-1,145,833  8,645,833-24,583,333
    Fluoride 3.1582 1,741-3,483 26,281-74,726
    Iodine 0.0197 279,188-558,376  4,213,198-11,979,695
    Manganese 0.5461 10,071-20,143 151,987-432,155
    Molybdenum 0.0078 705,128-1,410,256 10,641,026-30,256,410
    Selenium 0.0117 470,085-940,171  7,094,017-20,170,940
    Zinc 2.6002 2,115-4,230 31,921-90,762
  • The dry composition may also be available in a concentrated form. In some aspects, the present disclosure provides a concentrated composition comprising a dry composition described herein, and an initial aqueous solution. The concentrated composition described herein may be advantageous since it may minimize storage space, weight, and transport of water and may also increase the stability and thereby also the shelf-life of the dialysis components. The concentrated composition may be stable for long term storage and transportation. The concentrated composition may be available in a container, in which additional aqueous solution may be added to the container to bring the solution to volume, at which point, the concentrated composition may be ready to use by the subject. The container may have the capacity to hold approximately 1, 2, 2.5, 4, or 10 liter of total aqueous solution. The additional aqueous solution may be added to the concentrated composition prior to administration to a subject for dialysis.
  • Food Composition
  • In some aspects, the present disclosure provides a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least one nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline, wherein each nonessential amino acid is 0-1% wt/wt of protein of the food composition, wherein the food composition comprises 0-1% wt/wt of cysteine and/or cystine of protein of the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • In certain embodiments, the amount of each nonessential amino acid, selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline, is 0-2% wt/wt of the protein in the food composition. The amount of each nonessential amino acid may be approximately 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0% wt/wt of the protein. The amount of each nonessential amino acid may be 0-1% wt/wt of the protein in the food composition. In one embodiment, the amount of each nonessential amino acid is 0-0.1% wt/wt of the protein in the food composition. In some embodiments, the food composition comprises at least two of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least three of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least four of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least five of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. The food composition may comprise at least six of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, and proline. In some embodiments, at least one of the nonessential amino acid is glycine. In some embodiments, at least one of the nonessential amino acid is proline.
  • In certain embodiments, the amount of the cysteine and/or cystine is 0-2% wt/wt of the protein in the food composition. The amount of the cysteine and/or cystine may be 0-1% wt/wt of the protein in the food composition, such as 0, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1% wt/wt of the protein. In an exemplary embodiment, the amount of cysteine and/or cystine is 0-0.2% wt/wt of the protein in the food composition. In one embodiment, the amount of cysteine and/or cystine is approximately 0-0.1% wt/wt of the protein in the food composition. In another embodiment, the amount of cysteine and/or cysteine is 0-0.04% wt/wt of the food composition, such as approximately 0, 0.01, 0.02, 0.03, or 0.04% wt/wt of the food composition.
  • In some aspects, the present disclosure provides a food composition comprising: (a) a cysteine devoid protein source; (b) at least one essential amino acid; and (c) at least six nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine, wherein each nonessential amino acid is 0-1% wt/wt of protein in the food composition, and wherein the food composition comprises at least 6% wt/wt of protein.
  • In some aspects, the present disclosure provides a food composition comprising: (a) at least one essential amino acid; and (b) glutamate, wherein the food composition is substantially free of nonessential amino acids other than glutamate. In some aspects, the present disclosure provides a food composition comprising: (a) at least one essential amino acid; (b) glutamate; and (c) arginine, wherein the food composition is substantially free of nonessential amino acids other than glutamate.
  • In certain embodiments, the amount of each nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine is 0-2% wt/wt of the protein in the food composition. The amount of each nonessential amino acid may be approximately 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0% wt/wt of the protein. The amount of each nonessential amino acid may be 0-1% wt/wt of the protein in the food composition. In one embodiment, the amount of each nonessential amino acid is 0-0.1% wt/wt of the protein in the food composition.
  • In some embodiments, the food composition comprises at least seven of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least eight of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least nine of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least ten of the nonessential amino acid selected from a group consisting of alanine, asparagine, aspartate, glutamic acid, glycine, proline, glutamine, serine, tyrosine, and cysteine. The food composition may comprise at least six of the nonessential amino acids are selected from the group consisting of glycine, proline, glutamine, serine, tyrosine, and cysteine. In some embodiments, at least one of the nonessential amino acid is glycine. In some embodiments, at least one of the nonessential amino acid is proline. In an exemplary embodiment, at least one of the nonessential acids is cysteine. When the food composition comprises cysteine, the amount of cysteine and/or cystine is 0-2% wt/wt of protein in the food composition. The amount of the cysteine and/or cystine may be approximately 0-1% wt/wt of the protein in the food composition, such as approximately 0, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1% wt/wt of the protein. In an exemplary embodiment, the amount of cysteine and/or cystine is 0-0.2% wt/wt of the protein in the food composition. In one embodiment, the amount of cysteine and/or cystine is 0-0.1% wt/wt of the protein in the food composition. In another embodiment, the amount of cysteine and/or cysteine is 0-0.04% wt/wt of the food composition, such as approximately 0, 0.01, 0.02, 0.03, or 0.04% wt/wt of the food composition. In an exemplary embodiment, the food composition is substantially free of cysteine and/or cystine. The food composition may be free of cysteine and/or cystine.
  • Any food composition as described herein, in certain embodiments, the amount of the cysteine devoid protein source is at least 1% wt/wt of the food composition. The cysteine devoid protein source may be gelatin. The amount of the a cysteine devoid protein source may be 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, or 10.0% wt/wt of the food composition. In some embodiments, the amount of the cysteine devoid protein source is 1-5% of the food composition.
  • Any food composition described herein, in certain embodiments, the amount of protein is at least approximately 5% of the total weight of the food composition. The amount of protein in the food composition may be approximately 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25% of the total weight of the food composition. In one embodiment, the amount of protein is approximately 6% of the total weight of the food composition. In another embodiment, the amount of protein is approximately 16% of the total weight of the food composition.
  • Any food composition as described herein, in certain embodiments, the essential amino acid is selected from a group consisting of arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine. In an exemplary embodiment, the essential amino acid is methionine. In an exemplary embodiment, the essential amino acid is arginine. The essential amino acid may be approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, or 75% wt/wt of the food composition. In some embodiments, the essential amino acid is at least approximately 2% wt/wt of the food composition. The amount of the essential amino acid may be 2-30% wt/wt of the food composition. In an exemplary embodiment, the amount of the essential amino acid is approximately 8% wt/wt of the food composition.
  • In one embodiment, the food composition comprises at least two essential amino acids. The food composition may comprise at least three essential amino acids. The food composition may comprise at least four essential amino acids. The food composition may comprise at least five essential amino acids. The food composition may comprise at least six essential amino acids. The food composition may comprise at least seven essential amino acids. The food composition may comprise at least eight essential amino acids. The food composition may comprise at least nine essential amino acids. The food composition may comprise at least ten essential amino acids.
  • Any food composition described herein may further comprise at least one vitamin. The vitamin may be selected from a group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline. In an exemplary embodiment, the vitamin is vitamin C. In one embodiment, the food composition comprises at least approximately 10 mg of vitamin C per kg of the food composition. The food composition may comprise approximately 10, 15, 20, 25, 30, 40, 45, 50, 55, or 60 mg of vitamin C per kg of the food composition. In an exemplary embodiment, the food composition comprises approximately 40 mg of vitamin C per kg of the food composition.
  • In certain embodiments, any food compositions described herein further comprises an essential fatty acid, such as omega-6 and omega-3 fatty acids. In one embodiment, the essential fatty acid is an omega-3 fatty acid. The amount of essential fatty acid may be approximately 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 mg per liter. In some embodiments, the amount of essential amino acids is 1200-1600 mg per liter. In some embodiments, the amount of essential amino acids is 100-200 mg per liter.
  • In certain embodiments, any food compositions described herein further comprises at least one mineral. The mineral may be selected from a group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc. In an embodiment, the food composition comprises at least approximately 1% w/w of mineral, such as 1, 2, 3, 4, or 5% w/w of mineral in the food composition. The amount of at least one of the mineral may be 0.05 mg-20 g per kg of the food composition, such as approximately 0.1, 1, 10, 50, 100, 500, 1,000, 5,000, 10,000 or 20,000 mg of mineral per kg of the food composition.
  • Any food compositions described herein further comprise at least one ion. In certain embodiments, the amount of at least one ion is at least 0.1% w/w, such as 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0% w/w of ion in the food composition. The amount of at least one ion may be 0.05-20 g per kg food, such as approximately 0.05, 0.1, 1, 5, 10, 15, or 20 g of ion per kg of the food composition.
  • Any food compositions described herein further comprise glutamate. In some embodiments, the food composition comprises at least, approximately, or no more than 0.1 mg of glutamate, such as approximately 0.1, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 mg. The amount of glutamate may be 0.1-50 mg, 5-25 mg, or 25-45 mg.
  • In some embodiments, any of the food compositions described herein comprise at least approximately 10% wt/wt of amino acids, such as 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20% w/w or amino acids. In some embodiments, the food composition comprises approximately 14% wt/wt of amino acids. In some embodiments, the food composition comprises approximately 17% wt/wt of amino acids. In some embodiments, the food composition comprises 10-20% w/w of total amino acids. The food composition may comprise 14-17% w/w of total amino acids.
  • Any food composition as described herein, in certain embodiments, the food composition further comprises a pharmaceutically active agent. The amount of pharmaceutical agent, or any other component used in the food compositions described herein can be used in an amount that is therapeutically effective. The therapeutic amount can be assessed for individual subjects, or for groups of subjects. The group of subjects can be all potential subjects, or subjects having a particular characteristic such as age, weight, race, gender, or physical activity level. The pharmaceutically active agent may include anti-cancer therapeutics, such as mesna. The pharmaceutically active agent provided to the subject may be approximately 5, 10, 50, 100, 500, 1000, 1200, 1500, 2000, 2500, or 3000 mg. In some embodiments, the pharmaceutically active agent provided to the subject is 10-2000 mg. In some embodiments, the mesna is provided to the subject is 200-1200 mg. In some embodiments, the pharmaceutically active agent is an anti-obesity therapeutic.
  • Methods of Use
  • The invention provides for methods of treating (including preventing) cancer, metabolic diseases, such as obesity, and/or renal disease in subjects in need thereof. The method can include the reduction, depletion and/or supplementation of certain nutrients to the subject's diet. The method can include administering to the subject a food composition described herein that has no more than certain amounts of pre-selected nutrients. The nutrients that are reduced or depleted can be certain nutrients that are demanded at a higher level or degree by cancerous cells as compared to non-cancerous cells. The subject's diet can be controlled by administering to the subject a food composition wherein the subject does not eat any other foods or nutrients other than those that are part of the food composition, or permitted under the regimen. Therefore, the food composition may consist of all of the foods provided to the subject, and the food composition may have only certain amounts of pre-selected nutrients. The food composition can be designed such that the nutrients provided to or ingested by the subject are evaluated or determined on an hourly, daily, weekly or monthly basis. For any numbers provided herein that are described on a daily basis, the invention provides for proportionally scaled numbers for such amounts on an hourly, weekly or monthly basis. The food composition can be provided to the subject, alone or in combination with the dry composition, concentrated composition or liquid composition, until consistent and significant improvement is seen in the subject, and may be modified at any point of the treatment to fit the needs of the subject.
  • The methods described herein also include the reduction, depletion and/or supplementation of certain nutrients in the subject's system, such as blood levels, through dialysis. The present disclosure envisions methods of performing dialysis. Dialysis machines for performing dialysis, such as Freedom Cycler (Fresenius Medical Care), HomeChoice PRO™ (Baxter Healthcare Corporation), HomeChoice™ (Baxter Healthcare Corporation), Liberty® Cycler (Fresenius Medical Care), Newton™ IQ Cycler System (Fresenius Medical Care), 2008K@Home™ (Fresenius Medical Care), NxStage® System One™ with PureFlow™ SL (NxStage Medical, Inc.), Quanta SC+(Quanta Dialysis Technologies) and the like are known in the art, and may be used to carry out the methods described herein. The methods can include administering to the subject a liquid composition, concentrated composition, or dry composition described herein that has no more than certain pre-selected nutrients, which can be used alone or in combination with a diet that administers a food composition. In a preferred embodiment, the methods comprise placing a subject in need thereof on a nutritional diet with any of the food compositions disclosed herein and on dialysis with a dialysate composition (liquid composition, dry composition, or concentrated composition). In some embodiments, the subject is placed on a nutritional diet before, during, and/or after dialysis treatment. The nutrients that are reduced or depleted can be certain nutrients that are demanded at a higher level or degree by cancerous cells as compared to non-cancerous cells. The levels of these nutrients can be controlled by administering to the subject a dry composition, concentrated composition, or liquid composition and periodically monitoring the subject. For example, the methods may involve administering the composition to draw waste, fluid, and/or certain nutrients, such as cysteine and/or cystine, from the subject's blood. The dry composition, concentrated composition, or liquid composition may consist of a substantial amount of nutrients necessary for the subject, such as one or more essential amino acids, arginine, and glutamate. The methods may also involve administering the composition to provide the subject with necessary nutrients. The dialysate composition may be in the subject body as long necessary for the composition to exchange waste, fluids, and/or nutrients with the body. The exchange (drain and refill) may be facilitated by the subject or with the assistance of a machine. In some embodiments, the exchange takes at least 15, 20, 25, 30, 35, 40, 45, 50, 55, or 60 minutes.
  • The dry composition, concentrated composition or liquid composition can be designed such that the nutrients provided to the subject are evaluated or determined on an hourly, daily, weekly or monthly basis. For any numbers provided herein that are described on a daily basis, the invention provides for proportionally scaled numbers for such amounts on an hourly, weekly or monthly basis. The dry composition, concentrated composition, or liquid composition, alone or in combination with a food composition, can be provided to the subject until consistent and significant improvement is seen in the subject, and may be modified at any point of the treatment to fit the needs of the subject.
  • In some embodiments, the subject undergoes peritoneal dialysis. Peritoneal dialysis may involve the insertion of a flexible tube, such as a catheter, into the subject. The catheter may go inside the subject's abdomen or chest to outside the subject's body through an incision in the subject's skin. The catheter can fill the abdomen with dialysate FIG. 14 illustrates an exemplary dialysis system.
  • In some embodiments, the subject undergoes hemodialysis.
  • Treating Cancer
  • The methods described herein may comprise providing a composition described herein. In some aspects, the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • In some aspects, the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • In some aspects, the present disclosure provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • In certain embodiments, treating cancer is characterized by a reduction in tumor size. The tumor size may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial tumor size prior to treatment. The tumor may be reduced by at least 30%. The tumor may be reduced by at least 50%. The tumor may be reduced by at least 80%. The reduction of tumor size may be 10-100%. The reduction of tumor size may be 30-100%.
  • In certain embodiments, treating cancer is characterized by a reduction in circulated cancer cell counts. The circulated cancer cell count may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial circulated cancer cell count prior to treatment. The circulated cancer cell count may be reduced by at least 30%. The circulated cancer cell count may be reduced by at least 50%. The circulated cancer cell count may be reduced by at least 80%. The reduction of circulated cancer cell count may be 10-100%. The reduction of circulated cancer cell count may be 30-100%.
  • The methods of the present disclosure include monitoring levels of cysteine and/or cystine in the subject's blood. Monitoring cysteine and/or cystine may be used to modify the existing treatment in order to reduce the levels of cysteine and/or cystine to therapeutic levels.
  • In certain embodiments, treating cancer is characterized by decreased levels of plasma cysteine. The levels of plasma cysteine may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial levels of plasma cysteine prior to treatment. The levels of plasma cysteine may be reduced by at least 30%. The levels of plasma cysteine may be reduced by at least 50%. The levels of plasma cysteine may be reduced by at least 80%. The levels of plasma cysteine may be reduced by at least 90%. The levels of plasma cysteine may be reduced by at least 95%. The levels of plasma cysteine may be 30-100%.
  • Administering a composition described herein may result in a reduction of blood levels of cysteine and/or cystine. The composition may be administered in therapeutic amounts. Subsequent to the administration of a composition described herein, the level of cysteine in the subject's blood may be reduced to less than average levels of cysteine, such as less than approximately 10 μM of cysteine. In certain embodiments, the level of cysteine in the subject's blood is less than approximately 9 μM, 8 μM, 7 μM, 6 μM, 5 μM, 4 μM, 3 μM, 2 μM, or 1 μM In an exemplary embodiment, the level of cysteine in the subject's blood is approximately less than 1 μM. Subsequent to the administration of a composition described herein, the level of cystine in the subject's blood may be reduced to less than average levels of cystine, such as less than approximately 40 μM of cystine. In certain embodiments, the level of cystine in the subject's blood may be reduced to less than approximately 39 μM, 38 μM, 37 μM, 36 μM, 35 μM, 34 μM, 33 μM, 32 μM, 31 μM, 30 μM, 29 μM, 28 μM, 27 μM, 26 μM, 25 μM, 24 μM, 23 μM, 22 μM, 21 μM, 20 μM, 19 μM, 18 μM, 17 μM, 16 μM, 15 μM, 14 μM, 13 μM, 12 μM, 11 μM, 10 μM, 9 μM, 8 μM, 7 μM, 6 μM, 5 μM, 4 μM, 3 μM, 2 μM, or 1 μM. In an exemplary embodiment, the level of cystine in the subject's blood is approximately less than 5 μM.
  • Reducing Tumor Size
  • In some aspects, the present disclosure provides a method for reducing tumor size and/or circulated cancer cell counts in a subject in need thereof, the method comprising administering to the subject any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein.
  • In some aspects, the present disclosure provides a method for reducing tumor size and/or circulated cancer cell counts in a subject in need thereof, the method comprising administering to the subject a liquid composition of claim 1, a dry composition described herein, or a concentrated composition described herein.
  • In some aspects, the present disclosure provides a method for reducing tumor size and/or circulated cancer cell counts in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • Methods of the disclosure may result in suppressing tumor growth in a subject. In certain embodiments, the subject's tumor size does not increase by greater than 0-100%, such as approximately 0, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100%, as measured before and 6 weeks after the initial administration of any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein. The subject's tumor size may not increase by greater than approximately 100%, as measured before and 6 weeks after the initial administration of any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein. The subject's tumor size may not increase by greater than approximately 70%, as measured before and 6 weeks after the initial administration. The subject's tumor size may not increase by greater than approximately 60%, as measured before and 6 weeks after the initial administration. The subject's tumor size may not increase by greater than approximately 40%, as measured before and 6 weeks after the initial administration. The subject's tumor size may not increase by greater than approximately 20%, as measured before and 6 weeks after the initial administration. The subject's tumor size may not increase by greater than approximately 0%, as measured before and 6 weeks after the initial administration.
  • In certain embodiments, the subject's tumor size decreases by at least 10%, as measured before and 6 weeks after the initial administration of any food composition described herein, and administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition described herein. The subject's tumor size may decrease by at least 20%, as measured before and 6 weeks after the initial administration. The subject's tumor size may decrease by at least 30%, as measured before and 6 weeks after the initial administration. The subject's tumor size may decrease by at least 40%, as measured before and 6 weeks after the initial administration. The subject's tumor size may decrease by at least 50%, as measured before and 6 weeks after the initial administration.
  • Methods of the disclosure may result in a reduction in circulated cancer cell counts. The circulated cancer cell count may be reduced by at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100% of the initial circulated cancer cell count prior to treatment. The circulated cancer cell count may be reduced by at least 30%. The circulated cancer cell count may be reduced by at least 50%. The circulated cancer cell count may be reduced by at least 80%. The reduction of circulated cancer cell count may be 10-100%. The reduction of circulated cancer cell count may be 30-100%.
  • Treating Obesity
  • In some aspects, the present disclosure provides a method for treating obesity in a subject in need thereof, the method comprising administering to the subject any food composition described herein.
  • Treating obesity may characterized by weight loss in a subject. The subject may experience of at least or approximately 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50% weight loss from the start of the nutritional diet with the food compositions disclosed herein. In certain embodiments, the subject's weight loss is greater than 5%, as measured before and 6 weeks after commencement of the controlled diet. A controlled diet comprises administering a food composition described herein to a subject in need thereof. The subject's weight loss may be greater than 10%, as measured before and 6 weeks after commencement of the controlled diet. The subject's weight loss may be greater than 15%, as measured before and 6 weeks after commencement of the controlled diet. The subject's weight loss may be greater than 20%, as measured before and 6 weeks after commencement of the controlled diet. The subject's weight loss may be greater than 25%, as measured before and 6 weeks after commencement of the controlled diet. The subject's weight loss may be greater than 30%, as measured before and 6 weeks after commencement of the controlled diet.
  • Treating Renal Disease
  • In some aspects, the present disclosure provides a method for treating renal disease in a subject in need thereof, the method comprising administering to the subject a liquid composition described herein, a dry composition described herein, or a concentrated composition. The methods of the present disclosure may provide or deplete certain nutrients to the subject. The subject may be experiencing malnutrition, nutrition deficiency or other side effects from dialysis, and administration of a liquid composition, dry composition, or concentrated composition may prevent, alleviate, reduce, or eliminate such side effects. Other side effects may include, but are not limited to, low blood pressure, nausea, vomiting, dry and itchy skin, restless leg syndrome, muscle cramping, or combinations thereof. For the methods described herein administering a liquid composition or dry composition, in certain embodiments, the methods further comprise utilizing a membrane. The membrane may comprise modified cellulose diacetate or polysulfone. In an exemplary embodiment, the membrane is a low-flux membrane that permits small molecule exchange. In some embodiments, the low-flux membrane comprises a plurality of pores, in which the plurality of pores comprises a first end and a second end. In some embodiments, the low-flux membrane comprises a plurality of pores. The plurality of pores may have an average pore diameter of approximately 1.8 nm. In some embodiments, the low-flux membrane permits approximately 50% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. The low-flux membrane may permit approximately 75% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. In some embodiments, the low-flux membrane permits approximately 90% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. The low-flux membrane may permit approximately 95% or higher of a molecule weighing approximately 500 Daltons or less to move through the plurality of pores, from the first end to the second end. In some embodiments, approximately 20% of a molecule of at least 1000 Daltons passes through the plurality of pores, from the first end to the second end. Approximately 10% of a molecule of at least 1000 Daltons may pass from the first side to the second side. Approximately 1% of a molecule of at least 1000 Daltons may pass from the first side to the second side.
  • For any method described herein, the method can further comprise administering a pharmaceutically active agent. The pharmaceutically active agent may be an anti-cancer therapeutic. Examples of anti-cancer drugs include Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino-17-demethoxygeldanamycin, Alpharadin, Alvocidib, 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone, Amonafide, Anthracenedione, Anti-CD22 immunotoxins, Antineoplastic, Antitumorigenic herbs, Apaziquone, Atiprimod, Azathioprine, Belotecan, Bendamustine, BMW 2992, Biricodar, Brostallicin, Bryostatin, Buthionine sulfoximine, CBV (chemotherapy), Calyculin, cell-cycle nonspecific antineoplastic agents, Dichloroacetic acid, Discodermolide, Elsamitrucin, Enocitabine, Epothilone, Eribulin, Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE chemotherapy regimen, IT-101, Imexon, Imiquimod, Indolocarbazole, Irofulven, Laniquidar, Larotaxel, Lenalidomide, Lucanthone, Lurtotecan, Mafosfamide, Mitozolomide, Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1, Pawpaw, Pixantrone, Proteasome inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A, Sapacitabine, Stanford V, Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel, Triplatin tetranitrate, Tris(2-chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126 or Zosuquidar. The pharmaceutically active agent may be an anti-obesity therapeutic. Examples of anti-obesity therapeutic agents include lipase inhibitors (such as Orlistat), dopaminergic, noradrenergic, and serotoninergic compounds, cannabinoid receptor antagonists (such as rimonabant), exenatide, pramlintide, and CNS agents (such as topimerate). These examples are provided for discussion purposes only, and are intended to demonstrate the broad scope of applicability of the invention to a wide variety of drugs. It is not meant to limit the scope of the invention in any way.
  • In one embodiment, the pharmaceutically active agent is provided in a subtherapeutic amount. The amount of pharmaceutical agent, or any other component used in a combination composition described herein, can be used in an amount that is subtherapeutic. In some embodiments, using sub-therapeutic amounts of an agent or component can reduce the side effects of the agent. Use of subtherapeutic amounts can still be effective, particularly when used in synergy with other agents or components. A subtherapeutic amount of the agent or component can be such that it is an amount below which would be considered therapeutic. For example, FDA guidelines can suggest a specified level of dosing to treat a particular condition, and a subtherapeutic amount would be any level that is below the FDA suggested dosing level. The subtherapeutic amount can be about 1, 5, 10, 15, 20, 25, 30, 35, 50, 75, 90, or 95% less than the amount that is considered to be a therapeutic amount. The therapeutic amount can be assessed for individual subjects, or for groups of subjects. The group of subjects can be all potential subjects, or subjects having a particular characteristic such as age, weight, race, gender, or physical activity level.
  • For any method described herein, in certain embodiments, the methods can further comprise administering a nutritional supplement. In some embodiments, the nutritional supplement comprises one or more nutrients, which may include vitamins, minerals, water, carbohydrates, fats, proteins, and combinations thereof. Nutritional supplements may be packaged as vitamins, minerals, herbs, meal supplements, sports nutrition products, natural food supplements, and fortified food products. The nutritional supplement may comprise at least one amino acid selected from a group consisting of arginine, histidine, isoleucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine. The nutritional supplement may provide the subject with nutrients at levels in accordance with the Dietary Reference Intake (DRI), the Reference Daily Intake (RDI), the World Health Organization (WHO) guidelines, Recommended Dietary Allowances (RDAs), Adequate Intakes (Ais), and/or Daily Value (DV). The required levels of nutrients may be the same or different for adults, children, men, and women. In certain embodiments, the nutritional supplement comprises one or more nutrients selected from a group consisting of vitamin A, thiamine, riboflavin, niacin, pantothenic acid, vitamin B6, biotin, folate, cyanocobalamin, vitamin C, vitamin D, vitamin E, vitamin K, and choline. In certain embodiments, the nutritional supplement comprises one or more nutrients selected from a group consisting of calcium, chloride, chromium, copper, fluoride, iodine, iron, magnesium, manganese, molybdenum, phosphorus, potassium, selenium, sodium and zinc. In certain embodiments, the nutritional supplement comprises one or more nutrients selected from a group consisting of water, carbohydrates, protein, fiber, fat, cholesterol, and fatty acids.
  • Dosing, Treatment Regimen, and Administration
  • Dialysis Only
  • The present disclosure provides methods of providing dialysis therapies to subjects in need thereof. For the methods described herein administering a liquid composition, dry composition or concentrated composition described herein, the liquid composition, dry composition, or concentrated composition may be administered via traditional dialysis methods. For example, the compositions may be administered by hemodialysis, peritoneal dialysis, hemofiltration, hemodiafiltration, and intestinal dialysis. In an exemplary embodiment, the compositions are administered by hemodialysis or peritoneal dialysis (such as continuous ambulatory PD (CAPD) and continuous cycler-assisted PD (CCPD). The dialysis treatment may be used in combination with other conventional methods for treating cancer, obesity and renal disease, such as surgery, radiation therapy, immunotherapy, anti-cancer therapeutics, anti-obesity therapeutics, or renal disease therapeutics.
  • For the methods described herein administering a liquid composition, dry composition, or concentrated composition described herein, in certain embodiments, the composition is administered to the subject at least once a week. The composition may be administered to the subject at least three times a week. The composition may be administered to the subject at least five times a week. In some embodiments, the composition is administered to the subject over the course of at least one week. The composition may be administered to the subject over the course of at least four weeks. The composition may be administered to the subject over the course of at least 2 months. The composition may be administered to the subject over the course of at least 3 months.
  • In some embodiments, the dialysis may be provided to a subject in need thereof in a dialysis center, a hospital, a clinic, health care facility, at home, or any place that is clean and dry.
  • Diet Only
  • For the methods described herein any food composition described herein may be administered multiple times a week, such as 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more times a week. In certain embodiments, the food composition is administered to the subject at least once a week. The food composition may be administered to the subject at least three times a week. The food composition may be administered to the subject at least five times a week. In an embodiment, the food composition may be administered to the subject at least twenty-one times a week. In some embodiments, the food composition may be administered to the subject over the course of at least one week. The food composition may be administered to the subject over the course of at least four weeks. The food composition may be administered to the subject over the course of at least 2 months. The food composition may be administered to the subject over the course of at least 3 months.
  • Dialysis and Diet
  • In any of the methods described herein, in certain embodiments, the liquid composition, the dry composition or concentrated composition described herein is administered to the subject sequentially with any food composition described herein. In another embodiment, the liquid composition, the dry composition or concentrated composition described herein is administered to the subject simultaneously with any food composition described herein. In some embodiments, the food composition, and the liquid composition, dry composition or concentrated composition are administered to the subject at least once a week. The food composition, and the liquid composition, dry composition or concentrated composition may be administered to the subject at least three times a week. The food composition, and the liquid composition, dry composition or concentrated composition may be administered to the subject over the course of at least one week. The food composition, and the liquid composition, dry composition or concentrated composition may be administered to the subject over the course of at least four weeks.
  • Pharmaceutical Compositions and Formulations
  • Diet
  • Any food composition described herein may have a pre-selected caloric value. For example the food composition may comprise at least 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 1500 kcal. The food composition may have a caloric value 0-100, 0-200, 200-400, 400-600, 600-800, 800-1000 or 500-1000 kcal. The food composition may have a mass that is at least 5, 10, 50, 100, 200, 300, 400, 500, 600, 700, or 1000 grams. The food composition may have a mass that is 0-100, 100-200, 200-400, 400-600, or 100-500 grams.
  • Any food composition as described herein may be sold to a subject in a packaged container. In certain embodiments, the food composition is packaged as a solid food, semi-solid food, or beverage. The food composition may be a concoction of nutrient ingredients, an instant quick meal mix, a microwaveable frozen paste, a bake- or roast-ready dough, or a customizable mixture of several nutrient modules with flavor varieties. In some embodiments, the food compositions may be cooked, partially cooked, or uncooked. For example, the food composition may be baby formula, a soft drink, pulp-free juice, a nutritional shake, a granola bar, a dessert, or candy. In some embodiments, the food composition is fortified with vitamins, minerals, herbs, nutrients, and combinations thereof. The food composition may be packaged as a food product selected from a group consisting of a snack bar, cereal product, bakery product, and dairy product. For example, the food composition may be baby formula or baby food. The food composition may be a soft drink. The food composition may be a nutritional shake or nutritional snack, such as a granola bar. The food composition may be meal replacement drink or snack. The food composition may have a palatability enhancer. The palatability enhancer may be can be MSG, or some other form of glutamate, salt.
  • In some embodiments, the food composition is a pet food. The pet food can be a wet or dry pet food. The pet food can be formed as a kibble which may be coated. The coating may be a polymer coating that is applied to the kibble after the kibble is formed.
  • In any of the methods administering any food composition described herein, in certain embodiments, further comprises additives, fillers or excipients. Additives, fillers and excipients may be added to beverages, solid food, semi-solid food, and other consumable substances in the any food composition. Additives, fillers, and excipients may include acidulents, acidity regulators, anticaking agents, antifoaming and foaming agents, antioxidants, bulking agents, food coloring, color retention agents, emulsifiers, flavors, flavor enhancers, four treatment agents, glazing agents, humectants, tracer gas, preservatives, stabilizers, sweeteners, and thickeners. In an exemplary embodiment, additives, fillers and excipients include one or more corn starch, maltodextrin 10, cellulose, mineral mix S10001, sodium bicarbonate, vitamin mix V10001, and choline bitrartrate. In some embodiments, the additives are food coloring. Food coloring may include FD&C blue #1, FD&C blue #2, FD&C citrus red #2, FD&C green #3, FD&C red #3, FD&C red #40, FD&C yellow #5, and FD&C yellow #6. In an exemplary embodiment, dyes in the food composition include one or more FD&C red #40, FD&C blue #1, and FD&C yellow dye #5.
  • In any of the methods administering any food composition described herein, in certain embodiments, the methods further comprises preservatives. Preservatives may be added to beverages, solid food, semi-solid food, and other consumable substances in the food composition. Preservatives may include natural food preservatives, chemical food preservatives, or artificial preservatives. For example, preservatives may include acetic acid, ascorbic acid, calcium ascorbate, erythrobic acid, isoascorbic acid, potassium nitrate, potassium nitrite, sodium ascorbate, sodium erythorbate, sodium isoascorbate, sodium nitrate, sodium nitrite, wood smoke, benzoic acid, calcium sorbate, carnobacterium divergens M35, carnobacterium maltaromaticum CB1, ethyl lauroyl arginate, 4-hexylresorcinol, leuconostoc carnosum 4010, methyl-p-hydroxy benzoate, methyl paraben, potassium acetate, potassium benzoate, potassium bisulphite, potassium diacetate, potassium lactate, potassium metabisulphite, potassium sorbate, propyl-p-hydroxy benzoate, propyl paraben, sodium acetate, sodium benzoate, sodium bisulphite, sodium diacetate, sodium lactate, sodium metabisulphite, sodium salt of methyl-p-hydroxy benzoic acid, sodium salt of propyl-p-hydroxy benzoic acid, sodium sorbate, sodium sulphite, sodium dithionite, sorbic acid, sulphurous acid, calcium propionate, calcium sorbate, dimethyl dicarbonate, natamycin, potassium sorbate, propionic acid, sodium diacetate, sodium proprionate, socium sorbate, sorbic acid, ascorbic acid, ascorbyl palmitate, ascorbyl stearate, butylate hydroxyanisole, butylated hydroxytoluene, citric acid esters of mono- and di-glycerides, gum guaiacum, lecithin, lecithin citrate, monoglyceride citrate, monoisopropyl citrate, propyl gallate, sodium metabisulphite, tartaric acid, tertiary butyl hydroquinone, and tocopherols. In one embodiment, preservatives are selected from a group consisting of sulfites, vitamin E, vitamin C, and butylated hydroxytoluene. In certain embodiments, preservation of beverages, solid food, semi-solid food, and other consumable substances may be accomplished through physical preservation, such as through freezing, boiling, smoking, and dehydration.
  • Dialysis
  • In certain embodiments, the liquid composition is stored in a container. For example, the container may be a dialysis bag. In one embodiment, the dialysis bag is intended for single-use. The container may hold about 1500 to 5000 mL of liquid composition. In one embodiment, approximately 2000 mL of liquid composition is a single unit. In another embodiment, approximately 2500 mL of liquid composition is a single unit.
  • In certain embodiments, the dry composition or concentrated composition is stored in a container. For example, the container may be a dialysis bag. In one embodiment, the dialysis bag is intended for single-use. In some embodiments, the dry composition or concentrated composition further comprises an aqueous solution in the container, thereby producing a mixture. The aqueous solution may be a sterile or nonsterile fluid. In one embodiment, the container comprises approximately 2000 mL of aqueous solution. In another embodiment, the container comprises approximately 2500 mL of aqueous solution. In certain embodiments, the mixture has a pH of approximately 4.0-6.5. In certain embodiments, the mixture has an osmolarity of approximately 300-720 mOsmol/L.
  • Applications
  • Cancer
  • In any of the methods described herein for treating cancer, in certain embodiments, the present disclosure provides a method for treating cancer and preventing cancer with the food compositions and/or dialysis compositions disclosed herein. In some embodiments, the method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g., Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CIVIL), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-hodgkin lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Viral-Induced cancer. In some embodiments, the cancer is selected from a group consisting of leukemia, hematologic malignancy, solid tumor cancer, prostate cancer, breast cancer, liver cancer, or brain tumor. In an exemplary embodiment, the cancer is selected from a group consisting of lung cancer, liver cancer, breast cancer, prostate cancer, colon cancer, lymphoma, and leukemia.
  • In some aspects, the present disclosure provides a method of treating cancer in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • In some aspects, the present disclosure provides a method of depleting cysteine in a subject that has been placed on a low-cysteine diet, comprising: placing the subject under dialysis, wherein the dialysis reduces or depletes cysteine from the subject's body.
  • In some aspects, the present disclosure provides a method of treating cancer in a subject that has been placed on a dialysis procedure to reduce cysteine in the blood, comprising: placing the subject on a low-cysteine diet.
  • In some embodiments, the dialysis comprises administering a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein. In some embodiments, the low-cysteine diet is a food composition disclosed herein.
  • Obesity
  • In any of the methods described herein for treating obesity, in certain embodiments, the present disclosure provides a method for treating obesity and preventing obesity. In some embodiments, the method relates to the treatment or prevention of disorders that may result in obesity or be the cause of obesity such as overeating and bulimia, polycystic ovarian disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, Type II diabetics, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g., children with acute lymphoblastic leukemia. In some embodiments, the method relates to treating at least one obesity-related health condition, including, not limited to, cardiovascular diseases, type 2 diabetes, obstructive sleep apnea, cancer, osteoarthritis, and depression.
  • Renal Disease
  • In any of the methods described herein for treating renal disease, in certain embodiments, the present disclosure provides a method for treating renal disease and preventing renal disease. In exemplary embodiments, renal disease is chronic renal disease. In some embodiments, the method relates to treating a subject diagnosed with renal disorders as identified by medical history, physical findings and laboratory tests. For example, the subject may be diagnosed with acute or chronic renal failure, resulting from diabetes, ischemia, drug induced toxicity, post-transplantation rejection with or without the need for dialysis; glomerulonephritis; glomerulosclerosis; interstitial nephritis; and acute tubular necrosis. Subjects may have physical findings such as anuria, lethargy, coma and decreased general growth rate. Subject may have increased plasma levels of creatinine, urea and uric acid (BUN), proteinuria, decreased GFR, RPF and renal size as determined by urogram, altered acid/base balance and changes in urine specific gravity.
  • Subject
  • Cancer
  • In any of the methods described herein for treating cancer and/or reducing tumor size, in certain embodiments, the subject is a human. For example, the human subject may be an adult, a child, or an infant. In an exemplary embodiment, the human subject is a human adult. The human subject may be a human adult 18 years old or older. The human subject may be between the ages 18 and 90 years old. In a certain embodiment, the human subject is between 40 and 85 years old. In an exemplary embodiment, the human subject is between 65 and 80 years old.
  • In any of the methods described herein for treating cancer and/or reducing tumor size, in certain embodiments, the subject is an animal. The animal subject may be a domesticated animal, such as a dog or cat. The animal subject may be a mouse or rat. The animal subject may be a mouse or rat at least 7 weeks old. The animal subject may be a mouse or rat between 7 and 15 weeks old. The animal subject may be a mouse or rat between 10 and 11 weeks old.
  • Subjects that can be treated with a food composition, liquid composition, concentrated composition, and/or dry composition of the present disclosure include subjects that have been diagnosed as having acute myeloid leukemia, acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g., Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CIVIL), chronic myleoproliferative disorders colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-hodgkin lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Viral-Induced cancer, leukemia, hematologic malignancy, solid tumor cancer, prostate cancer, castration-resistant prostate cancer, breast cancer, Ewing's sarcoma, bone sarcoma, primary bone sarcoma, T-cell prolymphocyte leukemia, glioma, glioblastoma, hepatocellular carcinoma, liver cancer, or diabetes, related conditions, and combinations thereof. In some embodiments, the subjects treated with a food composition, liquid composition, and/or dry composition of the present disclosure include subjects that have been diagnosed as having a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, and prostate (e.g., benign prostatic hypertrophy (BPH).
  • In certain embodiments, the human subject has been diagnosed with leukemia, lung cancer, liver cancer, breast cancer, colon cancer, prostate cancer, stomach cancer, cervix uteri, esophagus, bladder, non-hodgkin lymphoma, and combinations thereof. In certain embodiments, the animal subject has been diagnosed with malignant lymphoma, skin cancer, mammary gland tumors, breast cancer, soft tissue sarcoma, bone cancer, and combinations thereof.
  • In certain embodiments, the subject has a genetic predisposition for cancer.
  • In certain embodiments, the subject has elevated cysteine biosynthetic gene expression levels.
  • Obesity
  • As used herein, the term “obesity” refers to a condition in which individuals are considered “overweight,” or “obese.” The term “obese” or “obesity” includes “extreme obesity,” “severe obesity,” and “super obesity.”
  • In any of the methods described herein for treating obesity, in certain embodiments, the subject is a human. For example, the human subject may be an adult or a child. In certain embodiments, the human subject is a human child. For example, a human child may be between the ages of 2 and 5 years old, the ages of 6 and 11 years old, and the ages of 12 and 17 years old. In certain embodiments, the human subject is a human adult. The human subject may be a human adult 18 years old or older. The human subject may be 18-90 years old. In a certain embodiment, the human subject is 30-80 years old. In an exemplary embodiment, the human subject is 45-65 years old.
  • In certain embodiments, the subject is an animal. The animal subject may be a domesticated animal, such as a dog or cat. The animal subject may be a mouse or rat. The animal subject may be a mouse or rat at least 7 weeks old. The animal subject may be a mouse or rat that is 7-15 weeks old. The animal subject may be a mouse or rat that is 10-11 weeks old.
  • Subjects that can be treated with a food composition, liquid composition, concentrated composition, and/or dry composition of the present disclosure include subjects that have been diagnosed as being overweight or obese and/or having a body mass index (BMI) of at least 25 kg/m2. Obesity in a subject can be assessed using a body mass index (BMI) measurement. A subject's BMI can be calculated by dividing the subject's body weight (in kg) by the square of the subject's height (m2). A subject can be considered obese if the subject exhibits a BMI that is over 25 kg/m2, over 26 kg/m2, over 27 kg/m2, over 28 kg/m2, over 29 kg/m2, over 30 kg/m2, over 31 kg/m2, over 32 kg/m2, over 33 kg/m2, over 34 kg/m2, over 35 kg/m2, over 36 kg/m2, over 37 kg/m2, over 38 kg/m2, over 39 kg/m2, over 40 kg/m2, over 41 kg/m2, over 42 kg/m2, over 43 kg/m2, over 44 kg/m2, over 45 kg/m2, over 46 kg/m2, over 47 kg/m2, over 48 kg/m2, over 49 kg/m2, or over 50 kg/m2. The subject may be diagnosed with class I obesity, class II obesity, class III obesity, or an equivalent thereof. The subject may have a BMI of at least 30 kg/m2, at least 35 kg/m2, at least 40 kg/m2, at least 45 kg/m2 or at least 50 kg/m2. The subject may have a BMI between 25 kg/m2 and 50 kg/m2. The subject may experience at least one obesity-related health condition, including, not limited to, cardiovascular diseases, type 2 diabetes, obstructive sleep apnea, cancer, osteoarthritis, and depression. A subject can also be considered to exhibit a propensity to develop NASH or hepatic steatosis if the subject exhibits abdominal obesity. Abdominal obesity can be assessed by measuring the circumference of the subject's waist. For example, if the subject is an adult male, the subject can be considered to exhibit abdominal obesity if the subject exhibits a waist circumference of 102 cm or greater. For other example, if the subject is an adult female, the subject can be considered to exhibit abdominal obesity if the subject exhibits a waist circumference of 88 cm or greater.
  • In certain embodiments, the subject habitually consumes an excessive amount of food, has a genetic predisposition for obesity, routinely lacks physical activity, has an adverse reaction to medication, has a mental disorder, has an endocrine disorder, and combinations thereof. In certain embodiments, the subject has a genetic predisposition for obesity. In certain embodiments, obesity in the subject is due to polymorphisms in one or more genes. In certain embodiments, the subject habitually consumes an excessive amount of food.
  • In certain embodiments, treating obesity is characterized by the subject losing at least 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 10% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 20% of the subject's initial body weight. Treating obesity may be characterized by the subject losing at least 30% of the subject's initial body weight.
  • Renal Disease
  • As used herein, the term “renal disease” includes, but is not limited to, kidney disease, nephropathy, nephritis, nephrosis, chronic kidney disease, chronic renal failure, acute renal disease and acute kidney injury.
  • In any of the methods described herein for treating renal disease, in certain embodiments, the subject is a human. For example, the human subject may be an adult or a child. In certain embodiments, the human subject is a human child. In certain embodiments, the human subject is a human adult. The human subject may be a human adult 18 years old or older. In certain embodiments, the human subject is at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 years old. The human subject may be at least 60 years old. The human subject may be between the ages 18 and 90 years old. In an exemplary embodiment, the human subject is between 55 and 75 years old.
  • In certain embodiments, the subject is an animal. The animal subject may be a domesticated animal, such as a dog or cat. The animal subject may be a mouse or rat. The animal subject may be a mouse or rat at least 7 weeks old. The animal subject may be a mouse or rat between 7 and 15 weeks old. The animal subject may be a mouse or rat that is 10-11 weeks old.
  • Subjects that can be treated with a food composition, liquid composition, and/or dry composition of the present disclosure include subjects that have been diagnosed with renal disease and/or a related condition. In certain embodiments, the subject has been previously or concurrently diagnosed with high blood pressure and/or diabetes, has an adverse reaction to medication, has a genetic predisposition for renal disease, experiences blockage to the urinary system, experiences inflammation (such as glomerulonephritis), and/or experiences infections (such as pyelonephritis).
  • Facilities
  • In some aspects, the present disclosure provides a facility comprising a plurality of liquid compositions. In certain embodiments, the facility manufactures the liquid composition described herein or components thereof. In one embodiment, the facility is a pharmaceutical manufacturing site. In certain embodiments, the facility distributes the liquid composition described herein or components thereof. For example, the facility may be a distribution center. In certain embodiments, the facility administers the liquid composition described herein or components thereof. In one embodiment, the facility is a hospital, dialysis center, home or clinic. The facility may administer the liquid composition to a subject through dialysis.
  • In some aspects, the present disclosure provides a facility comprising a plurality of dry compositions or concentrated compositions. In certain embodiments, the facility manufactures the dry composition or concentrated composition described herein or components thereof. In one embodiment, the facility is a pharmaceutical manufacturing site. In certain embodiments, the facility distributes the dry composition or concentrated composition described herein or components thereof. For example, the facility may be a distribution center. In certain embodiments, the facility administers the dry composition or concentrated composition described herein or components thereof. In one embodiment, the facility is a hospital or clinic. The facility may administer the dry composition or concentrated composition to a subject through dialysis.
  • In some aspects, the present disclosure provides a facility comprising a plurality of food compositions. In certain embodiments, the facility manufactures the food compositions described herein or components thereof. In one embodiment, the facility is a food manufacturing site. In certain embodiments, the facility distributes the food compositions described herein or components thereof. For example, the facility may be a distribution center. In certain embodiments, the facility provides the food compositions described herein and components thereof to the subject. In one embodiment, the facility may sell the food composition to the subject. In another embodiment, the facility may prepare the food composition. In another embodiment, the facility may administer the food composition to the subject. For example, the facility may be a distribution center, a commercial kitchen, a restaurant, an eating facility, or a medical facility. In some embodiments, the facility is a location that provides catering or food delivery service to hospitals, clinic or patients. The facility may be located in or within the vicinity of a hospital or clinic, such as within 0.1, 0.5, 1, or 5 miles of a hospital or clinic.
  • Kits
  • Diet
  • The invention also provides kits. In some aspects, the present disclosure provides a kit comprising: (a) a pre-packaged food composition as described herein; and (b) instructions for using the pre-packaged food composition. The kits include one or more food compositions described herein, in suitable packaging, and can further comprise written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like. The instructions may comprise an informational package describing the use and attendant benefits of the food composition in treating cancer, obesity or renal disease. Such kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the food composition, and/or which describe dosing, administration, the dieting regimen, side effects, drug interactions, or other information useful to the health care provider. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. A kit can comprise one or more complete meals and/or snacks of the food composition described herein. In some embodiments, a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more complete meals and/or snacks of the food composition. The kits may provide complete meals and/or snacks for exactly or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, or more days. Instructions for use can comprise diet regimen instructions, such as instructions to take 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more complete meals and/or snacks 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times per day. For example, a kit can comprise a complete meal and/or snack of the food composition supplied as a food product, with each food product packaged separately, multiples of a food product packaged separately according to the number of complete meals and/or snacks of the food composition per administration (e.g. pairs of components), or all food products packaged together (e.g. in a container). As a further example, a kit can comprise a complete meal and/or snack of the food composition as a bottled drink, the kit comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 24, 28, 36, 48, 72, or more bottles. In an alternative example, a kit can comprise a complete meal and/or snack of the food composition as a nutritional bar, the kit comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 24, 28, 36, 48, 72, or more nutritional bar. The kit may comprise components of the food composition provided or packaged as separate compositions in separate containers within the kit. In some embodiments, the kit may comprise components of the food composition provided or packaged as a single composition within a container of the kit.
  • The kit can further contain another agent. In some embodiments, the food composition or component thereof, and the agent are provided or packaged as separate compositions in separate containers within the kit. In some embodiments, the food composition or component thereof, and the agent are provided or packaged as a single composition within a container in the kit. Suitable packaging and additional articles for use (e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like) are known in the art and can be included in the kit. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits can also, in some embodiments, be marketed directly to the consumer.
  • The kit can further contain a pre-packaged liquid composition, pre-packaged concentrated composition, or a pre-packaged dry composition described herein. The kit can comprise instructions directing the administration of the pre-packaged liquid composition, or pre-packaged concentrated composition, or dry composition. In some embodiments, a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more pre-packaged liquid compositions, pre-packaged concentrated compositions or pre-packaged dry compositions.
  • In some embodiments, a kit can comprise a multi-day supply of complete meals and/or snacks of the food composition as described herein. The kit can comprise instructions directing the administration of the multi-day supply of complete meals and/or snacks of the food composition over a period of multiple days. The multi-day supply can be a one-month supply, a two-month supply, or a multi-week supply. The multi-day supply can be a 90-day, 180-day, 3-month or 6-month supply. The kit can include packaged daily complete meals and/or snacks of the food composition, such as packages of 1, 2, 3, 4, or 5 complete meals and/or snacks of the food composition. The kit can be packaged with other dietary supplements, vitamins, and/or meal replacement bars, mixes, and beverages.
  • Dialysis
  • In some aspects, the present disclosure provides a kit comprising: (a) a pre-packaged liquid composition as described herein; and (b) instructions for using the pre-packaged liquid composition. In some aspects, the present disclosure provides a kit comprising: (a) a pre-packaged dry composition or concentrated composition as described herein; and (b) instructions for using the pre-packaged dry composition. The kits include one or more liquid compositions, concentrated compositions, or dry compositions described herein, in suitable packaging, and can further comprise written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like. The instructions may comprise an informational package describing the use and attendant benefits of the liquid composition or dry composition in treating cancer, obesity or renal disease. Such kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the liquid composition, concentrated composition, or dry composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. A kit can comprise one or more dialysis bags containing the liquid composition, concentrated composition, or dry composition described herein. In some embodiments, a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more dialysis bags containing the liquid composition, concentrated composition, or dry composition. The kits may provide dialysis bags containing the liquid composition, concentrated composition, or dry composition for exactly or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, or more days. Instructions for use can comprise dialysis regimen instructions, such as instructions to use 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more dialysis bags containing the liquid composition, concentrated composition, or dry composition 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times per day. For example, a kit can comprise a dialysis bag containing the liquid composition, concentrated composition, or dry composition supplied as a single-use dialysis bag, with each single-use dialysis bag packaged separately, multiples of a single-use dialysis bag packaged separately according to the number of dialysis bags containing the liquid composition, concentrated composition, or dry composition per administration (e.g. pairs of single-use dialysis bag), or all single-use dialysis bag packaged together (e.g. in a container). The kit may comprise components of the dialysis bag containing the liquid composition, concentrated composition, or dry composition provided or packaged as separate compositions in separate containers within the kit. In some embodiments, the kit may comprise components of the dialysis bag containing the liquid composition, concentrated composition, or dry composition provided or packaged as a single composition within a container of the kit.
  • The kit can further contain another agent. In some embodiments, the dialysis bag containing the liquid composition, concentrated composition, or dry composition or component thereof, and the agent are provided or packaged as separate compositions in separate containers within the kit. In some embodiments, the liquid composition, concentrated composition, dry composition, or component thereof, and the agent are provided or packaged as a single composition within a container in the kit. Suitable packaging and additional articles for use (e.g., aqueous solution for reconstituting the dry composition or concentrated composition, measuring cup for liquid preparations, wrapping to minimize exposure to air, and the like) are known in the art and can be included in the kit. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits can also, in some embodiments, be marketed directly to the consumer.
  • The kit can further contain a pre-packaged liquid composition, pre-packaged concentrated composition, or a pre-packaged dry composition described herein. The kit can comprise instructions directing the administration of the pre-packaged liquid composition, pre-packaged concentrated composition or pre-packaged dry composition. In some embodiments, a kit comprises about, less than about, or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 31, 60, 90, 120, 150, 180, 210, or more pre-packaged liquid compositions, pre-packaged concentrated compositions or pre-packaged dry compositions.
  • In some embodiments, a kit can comprise a multi-day supply of dialysis bags containing the liquid composition, concentrated composition, or dry composition as described herein. The kit can comprise instructions directing the administration of the multi-day supply of dialysis bags containing the liquid composition, concentrated composition, or dry composition over a period of multiple days. The multi-day supply can be a one-month supply, a two-month supply, or a multi-week supply. The multi-day supply can be a 90-day, 180-day, 3-month or 6-month supply. The kit can include packaged dialysis bags containing the liquid composition, concentrated composition, or dry composition, such as packages of 1, 2, 3, 4, or 5 dialysis bags containing the liquid composition, concentrated composition, or dry composition. The kit can be packaged with a food composition described herein, dietary supplements, vitamins, and/or meal replacement bars, mixes, and beverages.
  • Systems
  • The present disclosure provides a system for placing a subject under dialysis and/or a nutritional diet with a composition disclosed herein to aid in the treatment or prevention of a disease. The system may carry out any of the methods disclosed herein and may use any of the compositions disclosed herein.
  • The dialysis systems of the present disclosure can be controlled and modified to provide various dialysis therapies, as desired. The dialysis systems may be modified (e.g. increase/decrease the number of exchanges, increase/decrease the amount of dialysate used during each exchange, and/or use an adjusted dialysate formulation) to adjust to a subject's medical progress. The dialysis systems may facilitate peritoneal dialysis, hemodialysis and other forms of dialysis. The dialysis systems may comprise (a) a dialysis container comprising a composition disclosed herein, (b) tubing, and (c) a collection container. In some embodiments, the dialysis systems further comprise a machine for facilitating and/or automating fluid exchange. In some aspects, the present disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein, and a dialysis machine configured to administer the liquid composition, the concentrated composition or the dry composition. In some embodiments, the dialysis machine comprises a pump, a pressure monitor, air trap, air detector, a clamp, and/or a filter. In some aspects, the present disclosure provides a dialysis system, comprising: a dialysis container comprising a liquid composition disclosed herein, a concentrated composition disclosed herein or a dry composition disclosed herein.
  • Peritoneal Dialysis.
  • Peritoneal dialysis (PD) is a treatment that uses the lining of a subject's abdomen, i.e. the peritoneum, and a dialysate to clean the subject's blood. Any of the liquid compositions, concentrated compositions, and dry compositions may be used as a dialysate in PD. Dialysate absorbs waste and fluid from the blood, using the peritoneum as a filter. In some embodiments, the dialysis system involves continuous ambulatory PD (CAPD) or continuous cycler-assisted PD (CCPD).
  • CAPD is “continuous,” machine-free and can be done at any time. The treatment may involve placing about two quarts of cleansing fluid into the subject's abdomen and later draining it. In some embodiments, CAPD can be done by hooking up a plastic bag of cleansing fluid to the tube in the peritoneum. The subject may raise the dialysis bag to approximately shoulder level to pull the dialysis fluid into the peritoneum. When empty, the dialysis bag is removed and discarded. When an exchange (putting in and taking out the fluid) is finished, the fluid is drained from the subject's abdomen. In some embodiments, this process is performed several times in a 24-hour period, such as 1, 2, 3, 4, 5, 6 or 7 times, or as needed. Each exchange may take about 30 minutes, 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, or 6 hours. FIG. 14A shows an exemplary CAPD process (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/peritoneal-dialysis). As shown in FIG. 14A, a patient undergoing CAPD may hooked up to a dialysis bag 101 and waste container 105 by a catheter placed in the patient's peritoneum and connecting tubes 104. The transfer set 103 connects the catheter to the dialysis bag 101 to perform dialysis exchange and can be designed to reduce the risk of infection. The transfer set may be “Y-shaped,” and one branch of the Y-tube may connect to the waste container 105, while the other branch may connect to a dialysis bag 101 containing a dialysis solution as described herein. In some embodiments, the dialysis solution is warmed prior to use by the subject. The dialysis bag 101 is placed above the patient's peritoneum in a way to allow the fluid in the dialysis bag to enter into the patient, such as on a pole as illustrated in FIG. 14A.
  • CCPD, also known as automated peritoneal dialysis (APD), differs from CAPD in that a machine (cycler) delivers and then drains the cleansing fluid for the subject. The treatment may be performed at any time, especially at night. The dialysis system automatically performs dialysis therapy on a patient, for example, during nighttime while the patient sleeps. The subject may be attached to the machine for an extended period of time, such as 8, 9, 10, 11, or 12 hours per day. FIG. 14B shows an exemplary CCPD process (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/peritoneal-dialysis). As shown in FIG. 14B, a patient undergoing CCPD may hooked up to a supply of dialysis solution disclosed herein 201 and waste line 205. The dialysis may be facilitated by a dialysis machine 204 (also known as a cycler). The dialysis machine may further comprise a heater 203. The flow of dialysis solution may be monitored by a fluid meter 202. The fluid meter 202 may measure and record how much solution the cycler 204 removes. In some embodiments, the cycler 204 compares the amount of dialysis solution that was put into the subject with the amount of fluid that drains out. The cycler 204 may to fill and empty the subject's peritoneum several times a night, such as three to five times, while the subject sleeps.
  • Hemodialysis.
  • Hemodialysis is a treatment to filter wastes and water from your blood. In hemodialysis, a dialysis machine and a special filter called an “artificial kidney,” or a “dialyzer,” are used to clean a subject's blood. The blood goes through the dialyzer, which is located outside of the subject's body. FIG. 15 is an exemplary schematic representation of hemodialysis (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/hemodialysis). FIG. 16 provides an overview of the filtering process in hemodialysis (source: https://www.niddk.nih.gov/health-information/kidney-disease/kidney-failure/hemodialysis). As shown in FIG. 15, a patient undergoing hemodialysis may be connected to a dialysis machine by inserting two needles in the subject's arm. Each needle is attached to a soft tube connected to the dialysis machine. The dialysis machine pumps blood through the filter and returns the blood to the patient. Through one of the needles, blood is removed for dialysis 307. At various stages of dialysis, the unfiltered and filtered blood is monitored with an arterial pressure monitor 308, dialyzer inflow pressure monitor 302, and venous pressure monitor 304. An anticoagulant 301 may be used to prevent blood clotting. The dialysis machine pumps the blood through a filter 303 (also known as a dialyzer) with a pump 309. The dialyzer 303 has two parts, one for your blood and one for a washing fluid called dialysate. A thin membrane separates these two parts. Once the blood enters the filter 303 at one end of the filter 401, the blood is forced into many, very thin, hollow fibers 402. As blood passes through the hollow fibers, dialysis solution 404 as disclosed herein passes in the opposite direction on the outside of the fibers. Waste products from the blood move into the dialysis solution and out of the filter 403. Filtered blood remains in the hollow fibers and returns to the body. Some nutrients from the dialysis solution may also return to the body with the filtered blood.
  • In some embodiments, hemodialysis is performed about 1, 2, 3, 4, 5, 6 or 7 times per week for about 1, 2, 3, 4, 5 or 6 hours at a time, or as needed.
  • EXAMPLES Example 1: Cancer Therapy Process
  • A cancer therapy regimen is provided to a patient previously identified as having a form of cancer. The patient is diagnosed with a tumor, solid or liquid, in the primary or metastatic stage. The regimen involves utilizing a combination of a nutritionally modified diet and dialysis with compositions disclosed here. The regimen involves metabolic intervention therapy that can be tailored to the needs of the individual patient. As depicted in FIG. 1, a metabolic cancer therapy can be provided to a patient diagnosed with cancer. The cancer patient is treated with dialysis and/or a nutritional diet in order to deplete targeted nutrient(s)
  • The patient is primed with any food composition described herein, i.e. a depleting diet, for approximately 3 days (“the priming stage”). During the priming stage, the patient's entire diet comprises any food composition described herein, with the exception of food and beverages approved by the regimen. The patient's plasma amino acid levels are measured and profiled by targeted LC-MS assay before and after the priming stage.
  • On approximately day 4 of the regimen, the patient continues to consume a depleting diet, and begins dialysis treatment—e.g. hemodialysis or peritoneal dialysis. The patient may undergo hemodialysis or peritoneal dialysis alone, or undergo either form dialysis serially or interchangeably within the therapy. A patient undergoing hemodialysis undergoes dialysis with a liquid composition, concentrated composition, or dry composition described herein (“dialysis solution”) at a facility described herein. The patient receives the dialysis solution for approximately 4 hours during the day or overnight. The patient receives dialysis treatment at least 3 times a week, for approximately 4 hours for each dialysis treatment. After each dialysis treatment, the patient's plasma amino acid profile is analyzed for monitoring treatment effect progression. A patient undergoing peritoneal dialysis undergoes dialysis with a liquid composition, concentrated composition, or dry composition described herein (“dialysis solution”) at a facility described herein, at the patient's home, or a convenient location. The patient receives the dialysis solution for approximately 8-12 hours per day or 24 hours continuously. For continuous peritoneal dialysis, the patient carries a portable device that administers the dialysis solution. The patient's plasma amino acid profile is analyzed approximately every 3 days for monitoring treatment effect progression.
  • For a typical dialysis treatment with a single-time/single-day clearance of 30% or higher, the patient's amino acid profile achieves therapeutic depletion levels after no more than approximately 8 weeks, as determined by the LC-MS assay. At this point, the patient discontinues dialysis treatment, but continues to take depleting diets until complete remission is achieved. Once the patient reaches complete remission, the patient remains on the depleting diet for approximately 2 weeks to stabilize complete remission, in which the patient is tested throughout.
  • Throughout the cancer therapy regimen, the patient's disease progression is monitored by companion test. Any serious complications that arise may be conveniently stopped or limited by allowing the patient to resume a normal diet. The cancer therapy regimen is resumed once the patient's condition is stabilized. The patient is permitted to continue to take painkillers or other cancer medicines as long as it does not interfere with the metabolic intervention treatment.
  • Example 2: Obesity Therapy Process
  • For a compatibility primer test, dietary intervention may be administrated for 1-4 weeks with companion plasma nutrient test to indicate the safety and potential efficacy in an individual patient. As described in Example 1, during the test, the patient will only eat and drink the prescribed food composition. For long term use, the diet regimen may be administrated as long as the subject remains obese.
  • Example 3: Renal Disease Therapy Process
  • With technical improvement on safety and clearance efficiency in our dialysis procedure, the renal disease patient may solely rely on dialysis for normal nutrient nourishment through the administration of the dry composition, concentrated composition, or liquid composition described herein. The dialysis procedure, as set forth in Example 1, is followed. For long term use, dialysis may be administrated as long as the subject's renal function remains impaired or lost.
  • Example 4: Cancer Therapy Process: Diet Only
  • For a compatibility primer test, dietary intervention may be administrated for 1-4 weeks with companion cancer progression test and plasma nutrient test to indicate the safety and potential efficacy in an individual patient. As described in Example 1, during the test, the patient will only eat and drink the prescribed food. For long term use, the patient on the diet regimen may be administrated to the subject as long as the tumor is not completely cured.
  • Example 5: Cancer Therapy Process: Dialysis only
  • With technical improvement on safety and clearance efficiency in our dialysis procedure, the cancer patient may solely rely on dialysis both for targeted depletion of cancer-addicted nutrients in plasma and for normal nutrient nourishment through the administration of the dry composition, concentrated composition, or liquid composition described herein. A dialysis only treatment is particularly applicable to late-stage gastrointestinal cancer because patients usually lose the ability to eat and rely solely on nutrient infusion. For long term use, dialysis may be administrated to the subject as long as the tumor is not completely cured.
  • Example 6: Dialysis Fluid Composition
  • Table 2 as presented below provides an exemplary formulation of the dialysis fluid—dry composition, concentrated composition, or liquid composition—described herein.
  • TABLE 2
    Dialysis fluid composition
    Dextrose (/L) mmole
    Hemodialysis only  5.5-11.0
    peritoneal dialysis only  83-236
    Osmolarity (mOsmol/L) 300-718
    pH 6.0 (4.0-6.5)
    mmole mEq
    Ionic Concentration (/L)
    Sodium 132 132
    Calcium 1.75 3.5
    Potassium 2 2
    Magnesium 0.25 0.5
    Chloride 96 96
    Buffer (L)
    HCO3 20-50 20-50
    Acetate 2.0-5.0 2.0-5.0
    Lactate 40 40
    Amino acid (/L) micromole MW microgram
    Alanine 347.5 89.0935 30.96
    Arginine 60.25 174.2017 10.50
    Asparagine 57.5 132.1184 7.60
    Aspartic acid 8 133.1032 1.06
    Beta-alanine 19.5 89.0935 1.74
    Cystine 40.25 240.3 9.67
    Glutamic acid 72 147.1299 10.59
    Glutamine 547.5 146.1451 80.01
    Glycine 212.5 75.0669 15.95
    Histidine 83.5 155.1552 12.96
    Isoleucine 79.75 131.1736 10.46
    Leucine 119 131.1736 15.61
    Lysine 195 146.1882 28.51
    Methionine 22.25 149.2124 3.32
    Phenylalanine 55.25 165.19 9.13
    Proline 222.5 115.131 25.62
    Serine 109.75 105.093 11.53
    Threonine 137.25 119.1197 16.35
    Tryptophan 66.5 204.2262 13.58
    Tyrosine 63.75 181.1894 11.55
    Valine 242.5 117.1469 28.41
    Vitamin (/L) micromole Average RDI
    Vitamin A 0.052 50 IU
    Vitamin C 7.57 1333 mcg
    Vitamin B6 1.48 250 mcg
    Vitamin E 0.90 0.43 IU
    Folate (Vit B9) 15.18 6.7 mcg
    Vitamin D 0.0044 67 IU
    Vitamin K 0.0038 1.7 mcg
    Thiamine 0.075 20 mcg
    Riboflavin 0.053 20 mcg
    Niacin 4.71 580 mcg
    Vitamin B12 0.000030 0.04 mcg
    Pantothenic acid 0.38 83 mcg
    Biotin 0.0021 0.50 mcg
    Choline 79.99 8333 mcg
    Mineral (/L) micromole MW microgram
    Iron 2.3279 55.845 130.00
    Copper 0.2360 63.546 15.00
    Chromium 0.0096 51.9961 0.50
    Fluoride 3.1582 18.9984 60.00
    Iodine 0.0197 126.90447 2.50
    Manganese 0.5461 54.938044 30.00
    Molybdenum 0.0078 95.94 0.75
    Selenium 0.0117 78.96 0.92
    Zinc 2.6002 65.38 170.00
  • Example 7: Food Composition
  • Table 3 as presented below provides an exemplary formulation of the food composition described herein.
  • TABLE 3
    Food composition
    gm % kcal %
    Protein
    17 17.6
    Carbohydrate 68.6 69.8
    Fat 5 11.6
    kcal/gm 3.87
    Ingredient (gm) gm kcal
    Gelatin 131 524
    Essential amino acids
    L-Arginine # 0 0
    L-Histidine-HCl-H2O 3 13
    L-Isoleucine 6 22
    L-Leucine 7 28
    L-Lysine-HCl 3 14
    L-Methionine 5 19
    L-Phenylalanine 4 18
    L-Threonine 5 19
    L-Tryptophan 2 8
    L-Valine 4 16
    Nonessential amino acids
    L-Alanine
    L-Asparagine-H2O
    L-Aspartate
    L-Cystine
    L-Glutamic Acid
    L-Glutamine
    Glycine
    L-Proline
    L-Serine
    L-Tyrosine
    Total L-Amino Acids
    Corn Starch 550.5 2202
    Maltodextrin 10 125 500
    Cellulose 50 0
    Corn Oil 50 450
    Mineral Mix(including calcium, 35 0
    chloride, chromium, cupric, ferric,
    iodate, magnesium, manganous,
    phosphate, potassium, selenite,
    sodium, sulfate, zinc)
    Sodium Bicarbonate 7.5 0
    Vitamin Mix (vitamin A, C, D, E 10 40
    and B family)
    Choline Bitrartrate 2 0
    Total 1000.1 3872
  • Example 8: Effect of Diet Only on Cancer
  • The effect of nutritional diet was tested in colon, breast, prostate and pancreatic cancer. The mice in the studies were placed one of the diets shown in FIG. 2. FIG. 3 shows an overview of the diet compositions used in the studies of the present disclosure and Table 5 of Example 14 further describes the diets. In the studies shown in FIG. 4 to FIG. 13B, the “complete diet” was the “designed control” of FIG. 3. In FIG. 2, MEAD201 and MEAD203 were depleted in 5 and 9 non-essential amino acids, respectively. MEAD202 was lacking 3 non-essential amino acids and insufficient 1 non-essential amino acid. Protein Y is natural protein casein, a common food ingredient to provide protein. Protein Z is soy protein. Protein X is collagen (gelatin).
  • Colon Cancer
  • Procedure. Athymic nude mice were implanted with HCT116 cells by subcutaneous injection. The mice were approximately 5-6 week old. The study had two groups: mice administered with a complete diet and mice administered with MEAD202. Each group had 5 mice. The mice were placed on the respective diet on day 7. The mice were fed ad libitum. The complete diet and MEAD 202 compositions are described in Table 5 of Example 14. The tumor sizes were monitored 3 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • Results. As shown in FIG. 4, the mice that were treated with MEAD202 exhibited a significant decrease in tumor volume in comparison to the mice that were treated with a complete diet. During the study, the mice treated with MEAD202 consistently had a lower tumor volume than the mice treated the complete diet. By day 25, the tumor volume for the MEAD202 mice were over 25% lower than the complete diet mice.
  • Triple Negative Breast Cancer
  • Procedure. In FIG. 5A, NOD-SCID mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study in FIG. 5A had two groups: mice administered with a complete diet and mice administered with treatment diets [MEAD202 (day 28-45) and MEAD203 (after day 45)], and the mice were placed on the respective diet on day 20. In FIG. 5B, athymic nude mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study in FIG. 5B had 3 groups: mice administered with a complete diet, mice administered with MEAD202, and mice administered with MEAD203, and the mice were place on the respective diet on day 7. Each group had 5 mice. The complete diet and treatment diet compositions are described in Table 5 of Example 14. The mice were fed ad libitum. The tumor sizes were monitored 3 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • Results. As shown in FIG. 5A and FIG. 5B, the mice that were treated with a nutritional diet exhibited a significant decrease in tumor volume than the mice treated with a complete diet. In the study in FIG. 5A, the mice treated with the treatment diets consistently had a lower tumor volume than the mice treated the complete diet. By day 65, the tumor volume for the treatment diets mice were approximately 60% lower than the complete diet mice. In the study in FIG. 5B, the mice treated with MEAD203 and MEAD202 consistently had a lower tumor volume than the mice treated the complete diet. By day 23, the tumor volume for both the MEAD203 and MEAD202 mice were over 25% lower than the complete diet mice.
  • Prostate Cancer
  • Procedure. Athymic nude mice were implanted with PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had two groups: mice administered with a complete diet and mice administered with MEAD203. Each group had 5 mice. The mice were placed on the respective diet treatment on day 5. The mice were fed ad libitum. The complete diet and MEAD 203 compositions are described in Table 5 of Example 14. The tumor sizes were monitored 3 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • Results. As shown in FIG. 6, the mice that were treated with MEAD203 exhibited a significant decrease in tumor volume in comparison to the mice that were treated with a complete diet. During the study, the mice treated with MEAD203 consistently had a lower tumor volume than the mice treated the complete diet. By day 46, the tumor volume for the MEAD203 mice were approximately 60% lower than the complete diet mice.
  • Pancreatic Cancer
  • Procedure. Athymic nude mice were implanted with MIAPaCa-2 cells by subcutaneous injection. The mice were 6-8 weeks old. The study had two groups: mice administered with a complete diet and mice administered with treatment diets [MEAD201 (day 0-24), MEAD202 (day 25-39) and MEAD203 (after day 39)]. Each group had 5 mice. The mice were placed on the respective diet on day 0. The mice were implanted 7 days before being placed on the respective diet. The complete diet and MEAD 202 compositions are described in Table 5 of Example 14. The mice were fed ad libitum. The tumor sizes were monitored 2 times weekly. Tumor tissues and plasma samples were collected. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • Results. As shown in FIG. 7, the mice that were treated with the treatment diets exhibited a significant decrease in tumor volume in comparison to the mice that were treated with a complete diet. During the study, the mice treated with the treatment diets on average had a lower tumor volume than the mice treated the complete diet. By day 50, the tumor volume for the treatment diet mice were over 40% lower than the complete diet mice.
  • Example 9: Diet and Dialysis
  • The effect of nutritional diet and dialysis was tested in glioblastoma. The mice in the studies were placed on one of the diets shown in FIG. 2 and/or placed on dialysis with one of the compositions shown in FIG. 8. In MEAD102, MEAD103 and MEAD1 μM, an 8-component vitamin cocktail, 10-component vitamin cocktail and 10-component vitamin cocktail were used, respectively. For MEAD102, the vitamin cocktail comprised: vitamin B6, folate (Vit B9), thiamine, riboflavin, vitamin B12, biotin, choline, and inositol. For MEAD103, the vitamin cocktail comprised the MEAD102 cocktail, and niacinamide and pantothenic-Calcium. The compositions for MEAD101, MEAD102, MEAD103 and MEAD104 are described below in Table 8.
  • TABLE 8
    Dialysis compositions
    MEAD101 MEAD102 MEAD103 MEAD104
    Dextrose 236 mM 236 mM 236 mM 236 mM
    Sodium chloride 132 mM 132 mM 132 mM 132 mM
    Sodium lactate
    40 mM 40 mM 40 mM 40 mM
    Calcium chloride 1.75 mM 1.75 mM 1.75 mM 1.75 mM
    Magnesium chloride 0.25 mM 0.25 mM 0.25 mM 0.25 mM
    μmole μmole μmole μmol
    Essential + Arg/Glu Amino acid (/L)
    Arginine 60.25 60.25 60.25
    Glutamate 140 140 140
    Histidine 83.5 83.5 83.5
    Isoleucine 79.75 79.75 79.75
    Leucine 119 119 119
    Lysine 195 195 195
    Methionine 22.25 22.25 22.25
    Phenylalanine 55.25 55.25 55.25
    Threonine 137.25 137.25 137.25
    Tryptophan 204.23 204.23 204.23
    Valine 242.5 242.5 242.5
    Vitamin (/L)
    Vitamin A 0.052 0.052 0.052
    Vitamin C 7.57 7.57 7.57
    Vitamin B6 1.48 1.48 1.48
    Vitamin E 0.90 0.90 0.90
    Folate (Vit B9) 15.18 15.18 15.18
    Vitamin D 0.0044 0.0044 0.0044
    Vitamin K 0.0038 0.0038 0.0038
    Thiamine 0.075 0.075 0.075
    Riboflavin 0.053 0.053 2.65
    Niacinamide 4.71 4.71
    Vitamin B12 0.000030 0.000030 0.000030
    Pantothenic-Ca 0.38 0.38
    Biotin 0.0020 0.0020 0.0020
    Choline 79.99 79.99 79.99
  • Procedure. Athymic nude mice were implanted with U87MG cells by subcutaneous injection. The mice were 6-8 weeks old. The study in FIG. 9 had 3 groups: mice administered with a complete diet, mice administered with MEAD203, and mice administered with MEAD 203 and placed on dialysis with MEAD101, MEAD102 and MEAD103 at various time points. Each treatment group had 1 mouse. The mice were placed on the respective treatment on day 5. The mice were implanted 7 days before being placed on the respective diet. The mice were fed ad libitum. For dialysis session, approximately 2 mL of dialysis solution was injected in the peritoneal space, and drawn out approximately 2 hours later. The tumor sizes were monitored daily. Arrows indicate the day and type of dialysis fluid were used. The study in FIG. 10 had 4 groups: mice administered a complete diet, mice administered with diet only, mice administered with dialysis only, and mice administered with diet and dialysis. Each group had 6 mice. The mice were placed on the respective treatment on day 1. Diet MEAD203 was used as the treatment diet. Designed Control was used as the control diet. The mice were implanted 22 days before being placed on the respective diets. The mice were fed ad libitum. MEAD103 was used for the dialysis. For each dialysis session, approximately 2 mL of fluid was injected in the peritoneal space, and drawn out approximately 2 hours later. Dialysis was performed on every week day. Mice were monitored every weekday.
  • Results. In FIG. 9, the mouse on the MEAD203 diet and dialysis experienced a reduction in tumor volume. The mice on the complete diet were euthanized at day 16 because the tumor volume was over 2000 mm3 for 3 consecutive measurements. Additionally, the mice were euthanized due to concerns of over ulceration and infection between mice. As shown in FIG. 10, the mice that were administered a treatment of diet+dialysis and the mice treated with diet only exhibited a reduction in tumor volume as compared to the mice on a complete diet and the mice on dialysis only. During the study, the diet+dialysis mice consistently had the lowest tumor volume in comparison to the other treatment groups. By day 12, the mice that were administered a treatment of diet+dialysis and the mice treated with diet only exhibited approximately a 40% reduction in tumor volume as compared to the mice on a complete diet and the mice on dialysis only.
  • Example 10: Body Weight
  • Procedure. In FIG. 11A athymic nude mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in Example 16. Each group had 5 mice. The mice were placed on respective diets on day 20. The complete diet and treatment diet compositions are described in Table 5 of Example 14. The mice were fed ad libitum. The body weights were monitored 3 times weekly. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. In FIG. 11B, athymic nude mice were implanted with PC-3 cells by subcutaneous injection. mice that were implanted with implanted PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 2 groups: mice administered with MEAD203, and mice administered with complete diet. Each group had 5 mice. The mice were placed on respective diets on day 5. The mice were fed ad libitum. The complete diet and MEAD 202 compositions are described in Table 5 of Example 14. The body weights were monitored 3 times weekly. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001.
  • Results. As shown in FIG. 11, the mice that were administered a treatment of MEAD203 and the mice treated with MEAD202 exhibited a reduction in body weight as compared to the mice on a complete diet and the mice on natural chow. By day 23, the mice that were administered MEAD203 and the mice treated with MEAD202 exhibited approximately a 15% reduction in body weight as compared to the mice on a complete diet and the mice on dialysis only. Body weight loss were relatively stabilize for the treatment diets.
  • Example 11: Blood Sugar
  • Procedure. In FIG. 12A athymic nude mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in Example 16. Each group had 5 mice. The mice were placed on the respective diet on day 20. The complete diet and treatment diet compositions are described in Table 5 of Example 14. The mice were fed ad libitum. The plasma samples were collected before treatment started and after treatment completed. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. In FIG. 12B, athymic nude mice were implanted with PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 2 groups: mice administered with MEAD203, and mice administered with complete diet. Each group had 5 mice. The mice were placed on the respective diet on day 5. The mice were fed ad libitum. The complete diet and MEAD 203 compositions are described in Table 5 of Example 14. The plasma samples were collected before treatment started and after treatment completed. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. Glucose in plasma was measured with Nova Max Glucose Test Strips.
  • Results. As shown in FIG. 12A and FIG. 12B, blood sugar is not increased in the treatment groups. In FIG. 12A, the post-dose glucose levels of the MEAD202 mice and MED203 mice were similar or lower than the pre-dose levels. In FIG. 12B, the post-dose glucose levels of the MEAD203 mice were similar to its pre-dose levels.
  • Example 12: Ketosis
  • Procedure. In FIG. 13A, athymic nude mice were implanted with MDA-MDB-231 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 4 groups: mice administered with MEAD203, mice administered with MEAD202, mice administered with complete diet, and mice administered with natural chow. Natural chow is described in Example 16. Each group had 5 mice. The mice were placed on the respective diet on day 20. The complete diet and treatment diet compositions are described in Table 5 of Example 14. The mice were fed ad libitum. The plasma samples were collected before treatment started and after treatment completed. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. In FIG. 13B, athymic nude mice were implanted with PC-3 cells by subcutaneous injection. The mice were 5-6 weeks old. The study had 2 groups: mice administered with MEAD203, and mice administered with complete diet. Each group had 5 mice. The mice were placed on the respective diet on day 5. The mice were placed on the respective diets 5 days after implantation. The mice were fed ad libitum. The complete diet and MEAD 203 compositions are described in Table 5 of Example 14. The plasma samples were collected before treatment started and after treatment completed. Tumor tissues and plasma samples were collected, and only plasma samples were analyzed for glucose and beta-keto body measurement. Significance is shown: * for P<0.05, ** for P<0.01, *** for P<0.001. Beta-ketone body in plasma was measured with Nova Max Ketone Test Strips.
  • Results. As shown in FIG. 13A and FIG. 13B, ketosis is not triggered by the administration of the treatment diets. In FIG. 13A, the post-dose β-ketone levels of the MEAD202 mice and MED203 mice were similar to the pre-dose levels. In FIG. 13B, the post-dose β-ketone levels of the MEAD203 mice were similar to its pre-dose levels.
  • Example 13: Dialysis Fluid Composition
  • Table 4 as presented below provides an exemplary formulation of the dialysis fluid—dry composition, concentrated composition, or liquid composition—described herein.
  • TABLE 4
    Dialysis fluid composition
    Dextrose 236 mM
    Sodium chloride 132 mM
    Sodium lactate
    40 mM
    Calcium chloride 1.75 mM
    Magnesium chloride 0.25 mM
    μmole
    Essential + Arg/Glu Amino acid (/L)
    Arginine 60.25
    Glutamate 140
    Histidine 83.5
    Isoleucine 79.75
    Leucine 119
    Lysine 195
    Methionine 22.25
    Phenylalanine 55.25
    Threonine 137.25
    Tryptophan 204.23
    Valine 242.5
    Vitamin (/L)
    Vitamin A 0.052
    Vitamin C 7.57
    Vitamin B6 1.48
    Vitamin E 0.90
    Folate (Vit B9) 15.18
    Vitamin D 0.0044
    Vitamin K 0.0038
    Thiamine 0.075
    Riboflavin 0.053
    Niacinamide 4.71
    Vitamin B12 0.000030
    Pantothenic-Ca 0.38
    Biotin 0.0020
    Choline 79.99
    Mineral (/L)
    Iron 2.33
    Copper 0.24
    Chromium 0.010
    Fluoride 3.16
    Iodine 0.020
    Manganese 0.55
    Molybdenum 0.0078
    Selenium 0.012
    Zinc 2.60
  • Example 14: Food Composition
  • Table 5 as presented below provides an exemplary formulation of a food composition described herein. The food compositions below were the diets used in studies above.
  • TABLE 5
    Food composition
    MEAD201 MEAD203 MEAD204 MEAD205 Complete Diet
    (gm/kg) (gm/kg) (gm/kg) (gm/kg) (gm/kg)
    Dietary Amount and Amount and Amount and Amount and Amount and
    Components Calories Calories Calories Calories Calories
    gm % kcal % gm % kcal % gm % kcal % gm % kcal % gm % kcal %
    Protein 17.0 17.7 17.0 17.7 17.0 17.7 17.0 17.7 17.0 17.7
    Carbohydrate 70.3 73.0 70.3 73.0 70.3 73.0 70.3 73.0 70.3 73.0
    Fat 5 11.7 5 11.7 5 11.7 5 11.7 5 11.7
    overall kcal/gm 3.9 3.9 3.9 3.9 3.9
    Ingredient (gm) gm kcal gm kcal gm kcal gm kcal gm kcal
    Gelatin 0 0 0 0 0 0 0 0 0 0
    Essential
    amino acids
    L-Arginine 10.7 43 15.4 62 12.3 49 0.0 0 10.0 40
    L-Histidine-HCl- 6.4 26 9.2 37 7.4 30 8.2 33 6.0 24
    H2O
    L-Isoleucine 8.6 34 12.3 49 9.9 39 11.0 44 8.0 32
    L-Leucine 12.9 51.4 18.5 74.0 14.8 59.1 16.4 65.7 12.0 48
    L-Lysine-HCl 15.0 60.0 21.6 86.3 17.2 69.0 19.2 76.7 14.0 56
    L-Methionine 10.0 40.0 10.0 40.0 10.0 40.0 10.0 40.0 6.0 24
    L-Phenylalanine 12.5 49.9 18.0 71.8 14.3 57.4 16.0 63.8 8.0 32
    L-Threonine 8.6 34.3 12.3 49.3 9.9 39.4 11.0 43.8 8.0 32
    L-Tryptophan 2.1 8.6 3.1 12.3 2.5 9.9 2.7 11.0 2.0 8
    L-Valine 8.6 34.3 12.3 49.3 9.9 39.4 11.0 43.8 8.0 32
    Nonessential
    amino acids
    L-Alanine 10.7 42.8 0.0 0.0 12.3 49.3 13.7 54.8 10.0 40
    L-Asparagine- 5.4 21.4 0.0 0.0 0.0 0.0 0.0 0.0 5.0 20
    H2O
    L-Aspartate 10.7 42.8 0.0 0.0 0.0 0.0 0.0 0.0 10.0 40
    L-Cystine 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 4.0 16
    L-Glutamic Acid 37.2 148.9 37.2 148.9 37.2 148.9 37.2 148.9 30.0 120
    L-Glutamine 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 5.0 20
    Glycine 10.7 42.8 0.0 0.0 12.3 49.3 13.7 54.8 10.0 40
    L-Proline 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 5.0 20
    L-Serine 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 5.0 20
    L-Tyrosine 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 4.0 16
    Total L-Amino 170.0 680.0 170.0 680.0 170.0 680.0 170.0 680.0 170.0 680.0
    Acids
    Corn Starch 468 1872 468 1872 468 1872 468 1872 468 1872
    Maltodextrin 10 125 500 125 500 125 500 125 500 125.0 500
    Sucrose 100 400 100 400 100 400 100 400 100.0 400
    Cellulose 50 0 50 0 50 0 50 0 50.0 0
    Soybean oil 40 360 40 360 40 360 40 360 40.0 360
    t- 0.008 0 0.008 0 0.008 0 0.008 0 0.008 0
    Butylhydroquinone
    Mineral Mix 35 0 35 0 35 0 35 0 35 0
    S10022M
    Vitamin Mix 10 40 10 40 10 40 10 40 10 40
    V10037
    Choline Bitartrate 2.5 0 2.5 0 2.5 0 2.5 0 2.5 0
    Total 1000.5 3852.0 1000.5 3852.0 1000.5 3852.0 1000.5 3852.0 1000.5 3852.0
  • Example 15: Reduction of Cysteine Uptake with Glutamate and Vitamins
  • The studies of the present disclosure showed that plasma glutamate was increased by about 80-100% in mice when on MEAD treatment diets. The increase in plasma glutamate may result in a reduction of cysteine update due to the effects on the xCT transporter. This could theoretically halve all cells' capability of cysteine uptake.
  • The studies of the present disclosure show that vitamins have an important function in the reduction of cysteine. Vitamins (especially vitamin B family) are important for dialysis and the nutritional diet disclosed herein. The vitamins may maintain appropriate physiology in normal cells. Vitamins are important for neural health and metabolism. Vitamins may also help normal cells synthesize the target nutrients when these nutrients are not provided through diet or depleted through dialysis. Vitamin C was not included for mouse studies because vitamin C (ascorbic acid) is not vitamin for mice because they can make it internatively. Vitamin C is expected to be important in treating humans. Table 6 below provides a few exemplary vitamins for use in the compositions described herein.
  • TABLE 6
    Vitamins
    Vitamin (/L) μmol
    Vitamin A 0.052
    Vitamin C 7.57
    Vitamin B6 1.48
    Vitamin E 0.90
    Folate (Vit B9) 15.18
    Vitamin D 0.0044
    Vitamin K 0.0038
    Thiamine 0.075
    Riboflavin 0.053
    Niacinamide 4.71
    Vitamin B12 0.000030
    Pantothenic-Ca 0.38
    Biotin 0.0020
    Choline 79.99
  • Example 16: Natural Chow Composition
  • TABLE 7
    Natural chow composition
    Nutrients2
    Protein, % 21.0
    Arginine, % 1.29
    Cystine, % 0.36
    Glycine, % 0.97
    Histidine, % 0.53
    Isoleucine, % 0.86
    Leucine, % 1.57
    Lysine, % 1.18
    Methionine, % 0.62
    Phenylalanine, % 0.91
    Tyrosine, % 0.60
    Threonine, % 0.78
    Tryptophan, % 0.24
    Valine, % 0.97
    Serine, % 0.98
    Aspartic Acid, % 2.19
    Glutamic Acid, % 4.18
    Alanine, % 1.19
    Proline, % 1.31
    Taurine, % 0.03
    Fat (ether extract), % 5.0
    Fat (acid hydrolysis), % 6.3
    Cholesterol, ppm 142
    Linoleic Acid, % 2.12
    Linolenic Acid, % 0.27
    Arachidonic Acid, % 0.01
    Omega-3 Fatty Acids, % 0.45
    Total Saturated Fatty Acids, % 0.78
    Total Monounsaturated 0.96
    Fatty Acids, %
    Fiber (Crude), % 4.6
    Neutral Detergent Fiber3, % 16.0
    Acid Detergent Fiber4, % 5.8
    Nitrogen-Free Extract 53.4
    (by difference), %
    Starch, % 28.2
    Glucose, % 0.19
    Fructose, % 0.24
    Sucrose, % 3.25
    Lactose, % 1.34
    Total Digestible Nutrients, % 75.1
    Gross Energy, kcal/gm 4.11
    Physiological Fuel Value5, 3.43
    kcal/gm
    Metabolizable Energy, kcal/gm 3.03
    Minerals
    Ash, % 5.9
    Calcium, % 0.81
    Phosphorus, % 0.64
    Phosphorus (non-phytate), % 0.34
    Potassium, % 1.10
    Magnesium, % 0.22
    Sulfur, % 0.33
    Sodium, % 0.30
    Chloride, % 0.52
    Fluorine, ppm 9.3
    Iron, ppm 185
    Zinc, ppm 89
    Manganese, ppm 84
    Copper, ppm 14
    Cobalt, ppm 0.71
    Iodine, ppm 0.97
    Chromium (added), ppm 0.01
    Selenium, ppm 0.37
    Vitamins
    Carotene, ppm 1.5
    Vitamin K, ppm 3.3
    Thiamin, ppm 17
    Riboflavin, ppm 8.0
    Niacin, ppm 85
    Pantothenic Acid, ppm 17
    Choline Chloride, ppm 2000
    Folic Acid, ppm 3.0
    Pyridoxine, ppm 9.6
    Biotin, ppm 0.30
    B12, mcg/kg 51
    Vitamin A, IU/gm 15
    Vitamin D3 (added), IU/gm 2.3
    Vitamin E, IU/kg 99
    Ascorbic Acid, mg/gm 0.00
    Calories provided by:
    Protein, % 24.517
    Fat (ether extract), % 13.134
    Carbohydrates, % 62.349
    Vitamins
    Carotene, ppm 1.5
    Vitamin K, ppm 3.3
    Thiamin, ppm 17
    Riboflavin, ppm 8.0
    Niacin, ppm 85
    Pantothenic Acid, ppm 17
    Choline Chloride, ppm 2000
    Folic Acid, ppm 3.0
    Pyridoxine, ppm 9.6
    Biotin, ppm 0.30
    B12, mcg/kg 51
    Vitamin A, IU/gm 15
    Vitamin D3 (added), IU/gm 2.3
    Vitamin E, IU/kg 99
    Ascorbic Acid, mg/gm 0.00
    Calories provided by:
    Protein, % 24.517
    Fat (ether extract), % 13.134
    Carbohydrates, % 62.349
    *Product Code
    1. Formulation based on calculated values from the latest ingredient analysis information. Since nutrient composition of natural ingredients varies and some nutrient loss will occur due to manufacturing processes, analysis will differ accordingly.
    2Nutrients expressed as percent of ration except where otherwise indicated. Moisture content is assumed to be 10.0% for the purpose of calculations.
    3NDF = approximately cellulose, hemi-cellulose and lignin.
    4ADF = approximately cellulose and lignin.
    5Physiological Fuel Value (kcal/gm) = Sum of decimal fractions of protein, fat and carbo-hydrate (use Nitrogen Free Extract) × 4, 9, 4 kcal/gm respectively.
  • While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (44)

1.-30. (canceled)
31. A composition comprising a mixture of amino acids, wherein the mixture comprises:
(a) one or more of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, or valine; and
(b) none, or one or more nonessential amino acids selected from alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline, and wherein when one or more nonessential amino acids are present in the mixture, the amount and ratio of nonessential amino acids to methionine present is in an amount wherein the composition is substantially free of at least one nonessential amino acid and wherein the composition is effective to treat cancer, renal disease, metabolic disease or obesity.
32. The composition of claim 31, wherein when the one or more nonessential amino acids are present in the mixture of amino acids, at least one of the nonessential amino acid is present in the mixture in an amount that provides a molar ratio of methionine of:
(i) a molar ratio of methionine to alanine that is greater than about 0.06;
(ii) a molar ratio of methionine to asparagine that is greater than about 0.3;
(iii) a molar ratio of methionine to aspartate that is greater than about 2;
(iv) a molar ratio of methionine to cysteine or cystine that is greater than about 0.5;
(v) a molar ratio of methionine to glutamine that is greater than about 0.04;
(vi) a molar ratio of methionine to glycine that is greater than about 0.1;
(vii) a molar ratio of methionine to serine that is greater than about 0.2;
(viii) a molar ratio of methionine to tyrosine that is greater than about 0.3; or
(viiii) a molar ratio of methionine to proline that is greater than about 0.1.
33. The composition of claim 31, wherein the composition decreases the level of plasma cysteine in a subject after consumption of the composition compared to the level of plasma cysteine in the subject before consumption of the composition.
34. The composition of claim 33, wherein the level of plasma cysteine is reduced by at least 30%.
35. The composition of claim 31, wherein the mixture of amino acids further comprises arginine.
36. The composition of claim 35, wherein the amount of methionine is in a molar ratio of methionine to arginine that is greater than about 0.3.
37. The composition of claim 31, wherein the mixture of amino acids further comprises glutamate.
38. The composition of claim 37, when the amount of methionine is in a molar ratio of methionine to glutamate that is greater than about 0.3.
39. The composition of claim 31, further comprising gelatin.
40. The composition of claim 31, further comprising at least one vitamin selected from the group consisting of vitamin A, vitamin B, vitamin C, vitamin D, vitamin E, vitamin K, and choline.
41. The composition of claim 40, wherein the at least one vitamin comprises vitamin B or vitamin C.
42. The composition of claim 31, further comprising at least one ion selected from a group consisting of sodium, calcium, magnesium, chloride, and lactate.
43. The composition of claim 31, further comprising maltodextrin.
44. The composition of claim 31, further comprising at least one mineral selected from the group consisting of iron, copper, chromium, fluoride, iodine, manganese, molybdenum, selenium, and zinc.
45. The composition of claim 31, further comprising one or more preservatives.
46. The composition of claim 45, wherein the one or more preservatives is a citric acid ester of mono- and di-glycerides, lecithin citrate, monoglyceride citrate, or monoisopropyl citrate.
47. The composition of claim 31, further comprising a fatty acid.
48. The composition of claim 47, wherein the fatty acid is omega-3 fatty acid.
49. The composition of claim 31, comprising at least one or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
50. The composition of claim 31, comprising at least two or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
51. The composition of claim 31, comprising at least three or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
52. The composition of claim 31, comprising at least four or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
53. The composition of claim 31, comprising at least five or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
54. The composition of claim 31, comprising at least six or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
55. The composition of claim 31, comprising at least seven or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
56. The composition of claim 31, comprising at least eight or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
57. The composition of claim 31, comprising at least nine or more nonessential amino acids selected from the group consisting of alanine, asparagine, aspartate, cysteine or cystine, glutamine, glycine, serine, tyrosine or proline.
58. The composition of claim 31, wherein the composition is a solid food, a semi-solid food, or a liquid.
59. The composition of claim 58, wherein the composition is packaged as a food product, wherein the food product is a snack bar, a cereal product, a bakery product, a dairy product, or a nutritional shake.
60. The composition of claim 31, wherein one or more of alanine, asparagine, aspartate, cysteine or cystine, glutamine glycine, serine, tyrosine or proline are not present in the composition.
61. The composition of claim 31, wherein the one or more of histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, or valine are present in the composition in an amount sufficient to cause a reduction in tumor size in a subject with cancer.
62. The composition of claim 31, further comprising a therapeutic agent.
63. A method of treating cancer in a subject, the method comprising administering a therapeutically effective amount of the composition according to claim 31 to a subject in need of said treatment.
64. The method of treating cancer according to claim 63, wherein said treatment suppresses growth of a tumor in said subject.
65. The method of suppressing growth of a tumor according to claim 63, wherein the method is effective for reducing the size of a tumor in the subject.
66. The method of claim 63, wherein the subject has a genetic predisposition for cancer.
67. The method of claim 63, wherein the size of the tumor is reduced by at least 10%.
68. The method of claim 63, wherein the subject has liver cancer, breast cancer, lung cancer, leukemia, prostate, or colon cancer.
69. The method of claim 63, wherein the tumor does not increase in size by at least 10% after administration of the composition compared to the size of the tumor before administration.
70. A method of decreasing plasma cysteine levels in a subject, the method comprising administering to the subject the composition of claim 31, thereby decreasing plasma cysteine levels in the subject.
71. The method of claim 70, wherein the plasma cysteine levels in the subject are decrease by at least 10%.
72. The method of claim 70, wherein the subject has a genetic predisposition for cancer.
73. The method of claim 70, wherein the subject has liver cancer, breast cancer, lung cancer, leukemia, prostate, or colon cancer.
US17/552,660 2017-12-11 2021-12-16 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy Abandoned US20220354812A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/552,660 US20220354812A1 (en) 2017-12-11 2021-12-16 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762597392P 2017-12-11 2017-12-11
PCT/US2018/065045 WO2019118519A1 (en) 2017-12-11 2018-12-11 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy
US16/897,152 US11241407B2 (en) 2017-12-11 2020-06-09 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy
US17/552,660 US20220354812A1 (en) 2017-12-11 2021-12-16 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/897,152 Continuation US11241407B2 (en) 2017-12-11 2020-06-09 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Publications (1)

Publication Number Publication Date
US20220354812A1 true US20220354812A1 (en) 2022-11-10

Family

ID=66820620

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/897,152 Active US11241407B2 (en) 2017-12-11 2020-06-09 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy
US17/552,660 Abandoned US20220354812A1 (en) 2017-12-11 2021-12-16 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/897,152 Active US11241407B2 (en) 2017-12-11 2020-06-09 Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Country Status (7)

Country Link
US (2) US11241407B2 (en)
EP (1) EP3723772A4 (en)
JP (2) JP7428655B2 (en)
CN (1) CN111683668A (en)
AU (1) AU2018383715A1 (en)
CA (1) CA3084319A1 (en)
WO (1) WO2019118519A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3352586A4 (en) 2015-09-21 2019-05-15 The Board Of Trustees Of The Leland Stanford Junior University Nutritional treatment for cancer
WO2019118519A1 (en) 2017-12-11 2019-06-20 Filtricine, Inc. Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy
WO2020092616A1 (en) * 2018-10-31 2020-05-07 Rejuvenation Therapeutics Compositions useful in the treatment of hyperhomocysteinemia
CA3143834A1 (en) * 2019-07-19 2021-01-28 Xiyan LI Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy and other uses
CN111544449A (en) * 2020-05-29 2020-08-18 甘肃锦东医疗器械有限公司 Citric acid hemodialysis concentrate for supplementing vitamin D
CN111481570A (en) * 2020-05-29 2020-08-04 甘肃锦东医疗器械有限公司 Citric acid hemodialysis concentrate for supplementing vitamin K
US20220305178A1 (en) * 2021-03-24 2022-09-29 Yuanheng Biomedical Co., Ltd. Cancer treatment by in vitro amino acid deprivation
CN114947139A (en) * 2022-04-28 2022-08-30 成都尚医信息科技有限公司 Amino acid composition capable of influencing tumor growth, functional food and application of amino acid composition

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5895652A (en) * 1996-07-29 1999-04-20 Longevity Institute International Method of metabolic adjuvanation and cellular repair
US8012926B2 (en) * 2003-05-30 2011-09-06 Riken Amino acid composition and supplementary liquid containing the same
US8703725B2 (en) * 2002-09-20 2014-04-22 Nestec S.A. Nutritional compositions
US8778410B2 (en) * 2008-02-19 2014-07-15 Earnest Medicine Co., Ltd. Oral or enteral composition useful for recovery of physical functions
US11241407B2 (en) * 2017-12-11 2022-02-08 Filtricine, Inc. Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3024167A (en) * 1959-02-06 1962-03-06 Armour & Company Of Delaware Lyophilized protein hydrolysate suitable for parenteral administration
GB1263336A (en) * 1968-02-14 1972-02-09 Morton Norwich Products Inc Food composition
US3697287A (en) * 1969-01-28 1972-10-10 Morton Norwich Products Inc Amino acid food composition
US6020007A (en) * 1984-06-22 2000-02-01 Btg International Limited Fluid therapy with l-lactate and/or pyruvate anions
US4871550A (en) * 1986-09-05 1989-10-03 Millman Phillip L Nutrient composition for athletes and method of making and using the same
US4988724A (en) 1988-12-09 1991-01-29 Board Of Regents, The University Of Texas System Methods and improved formulations for the determination and treatment of malignant disease in patients
US5108767A (en) * 1991-06-10 1992-04-28 Abbott Laboratories Liquid nutritional product for persons receiving renal dialysis
DE69332959T2 (en) * 1992-02-04 2004-05-13 Baxter International Inc., Deerfield Peritoneal dialysis solutions and their use to reduce damage to mesothelial cells
AU678201B2 (en) * 1992-12-22 1997-05-22 Baxter International Inc. Improved amino acid solutions for treatment of peritoneal dialysis patients
DK0802257T3 (en) 1996-04-19 2002-10-28 Nestle Sa Immortalized cell line from human colon epithelial cells
EP0958832B1 (en) * 1998-05-21 2006-01-25 Nipro Corporation Albumin containing peritoneal dialysis fluid
US6274103B1 (en) * 1999-03-26 2001-08-14 Prismedical Corporation Apparatus and method for preparation of a peritoneal dialysis solution
PT1062879E (en) * 1999-06-26 2003-12-31 Braun Melsungen Ag AQUOSA SOLUTION FOR PARENTERAL NUTRITION
US8642581B1 (en) 2000-02-11 2014-02-04 Brian D. Halevie-Goldman Compositions and methods for the production of S-adenosylmethionine within the body
US20080221497A1 (en) * 2004-04-26 2008-09-11 Haik Jr George M Use of Arginine and Like Substances and methylglyoxal and Like Substances in Dialysis Machines
JP2009515948A (en) * 2005-11-18 2009-04-16 レノビズ・カンパニー・リミテッド Corrosion-preventing coating agent composition containing organoclay and method for producing the same
AU2007357726A1 (en) * 2007-08-15 2009-02-19 Cheiron Japan Co. Peritoneal dialysate
EP2344150A2 (en) 2008-09-19 2011-07-20 Nestec S.A. Nutritional support to prevent and/or mitigate bone marrow toxicity from a cancerous tumor
CA2737619A1 (en) 2008-09-19 2010-03-25 Nestec S.A. Nutritional support of the immune system during anti-cancer treatment
CA2653011A1 (en) * 2008-12-03 2010-06-03 Apotex Technologies Inc Method and apparatus for packaging and dispensing pharmaceuticals
PT2374452T (en) * 2008-12-05 2019-10-31 Ajinomoto Kk Nutrient composition
EP2521559A4 (en) * 2010-01-04 2013-05-01 Pentec Health Inc Nutritive compositions and methods of using same
CN101912394B (en) * 2010-08-06 2013-11-06 岳茂兴 Medicinal composition for treating coagulopathy haemorrhage and application thereof
ES2396650B2 (en) * 2011-07-21 2013-07-16 Universidad Complutense De Madrid USE OF A COMPOSITION IN THE DEVELOPMENT OF A DIALYSIS SOLUTION FOR THE TREATMENT OF CEREBROVASCULAR DISEASES BY PERITONEAL DIALYSIS.
WO2014197533A2 (en) 2013-06-05 2014-12-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Palatable foods for a methionine-restricted diet
CA2922557A1 (en) 2013-08-29 2015-03-05 Board Of Regents, The University Of Texas System Engineered primate cystine/cysteine degrading enzymes as antineogenic agents
CN103652926A (en) * 2013-12-30 2014-03-26 上海仙普生物科技有限公司 Food composition for tumor patients
US9724319B2 (en) 2014-02-27 2017-08-08 Nusirt Sciences, Inc. Compositions and methods for the reduction or prevention of hepatic steatosis
US20160278415A1 (en) * 2015-01-30 2016-09-29 Nutrient Foods, Llc Food Compositions Containing All Essential Nutrients
EP3352586A4 (en) 2015-09-21 2019-05-15 The Board Of Trustees Of The Leland Stanford Junior University Nutritional treatment for cancer
JP2019507754A (en) 2016-02-23 2019-03-22 キャンサー・リサーチ・テクノロジー・リミテッドCancer Research Technology Limited Dietary products lacking at least two nonessential amino acids
CA3143834A1 (en) 2019-07-19 2021-01-28 Xiyan LI Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy and other uses

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5895652A (en) * 1996-07-29 1999-04-20 Longevity Institute International Method of metabolic adjuvanation and cellular repair
US8703725B2 (en) * 2002-09-20 2014-04-22 Nestec S.A. Nutritional compositions
US8012926B2 (en) * 2003-05-30 2011-09-06 Riken Amino acid composition and supplementary liquid containing the same
US8778410B2 (en) * 2008-02-19 2014-07-15 Earnest Medicine Co., Ltd. Oral or enteral composition useful for recovery of physical functions
US11241407B2 (en) * 2017-12-11 2022-02-08 Filtricine, Inc. Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
F.S. Dioguardi ("Clinical use of amino acids as dietary supplement: pros and cons," J Cachexia Sarcopenia Muscle (2011) 2: 75–80. (Year: 2011) *
Metabolic Maintenance (downloaded 11-25-2023 from https://www.metabolicmaintenance.com/ Amino- Acids/L-Phenylalanine/; available online 4-15-2014. (Year: 2014) *
Protein Works. "What is Maltodextrin?", downloaded 11-25-2023 from https://www.theproteinworks.com/thelockerroom/what-is-maltodextrin/; available on the internet 6/25/2013 (Year: 2013) *

Also Published As

Publication number Publication date
JP2021505677A (en) 2021-02-18
EP3723772A1 (en) 2020-10-21
WO2019118519A1 (en) 2019-06-20
EP3723772A4 (en) 2021-09-08
JP2023174954A (en) 2023-12-08
JP7428655B2 (en) 2024-02-06
AU2018383715A1 (en) 2020-06-18
CN111683668A (en) 2020-09-18
CA3084319A1 (en) 2019-06-20
US11241407B2 (en) 2022-02-08
US20200297679A1 (en) 2020-09-24

Similar Documents

Publication Publication Date Title
US11241407B2 (en) Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy
US20220400730A1 (en) Compositions, methods, kits and systems for cancer treatment and metabolic intervention therapy and other uses
Singh Nutritional management of patients with urea cycle disorders
Van der Meulen et al. Effect of source of starch on net portal flux of glucose, lactate, volatile fatty acids and amino acids in the pig
Adamkin et al. Current trends and future challenges in neonatal parenteral nutrition
US20090017167A1 (en) Mixture and beverage made therefrom for protecting cellular hydration
CN103974701A (en) Improving / prophylactic agent for chronic kidney disease
Stevenson et al. Nutrition in R enal S upportive C are: Patient‐driven and flexible
Rolandelli et al. Nutritional support in the frail elderly surgical patient
ES2485827T3 (en) Enteral feeding by very caloric probe, rich in protein
Andel et al. Impact of early high caloric duodenal feeding on the oxygen balance of the splanchnic region after severe burn injury
Stratton et al. Role of enteral and parenteral nutrition in the patient with gastrointestinal and liver disease
Nakagawa et al. Copper-deficiency anemia after esophagectomy: A pitfall of postoperative enteral nutrition through jejunostomy
US20040137080A1 (en) Multivitamin regimen for renal patients
CN103690561B (en) Preparation method of oral liquid
US20220117943A1 (en) Formulations for personalized methods of treatment
Peterson et al. Systemic approach to parenteral nutrition in the ICU
Tonozzi Nutritional status
CN104147003A (en) Medicine composition of compound amino acid injection 17AA-I and application thereof
WO2015137383A1 (en) Chemotherapy adjuvant
Callan et al. Parenteral Nutrition in paediatrics
Fuhrman Nutrition support for oncology patients
Zhang et al. Research progress on nutritional support in the neonatal and pediatric populations receiving extracorporeal membrane oxygenation
Mauldin Nutrition Alterations and Management
MacDonald Principles of Dietary Management

Legal Events

Date Code Title Description
AS Assignment

Owner name: FILTRICINE, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, XIYAN;WANG, XIN;YANG, XIAOLU;REEL/FRAME:058800/0965

Effective date: 20211217

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