US20150118306A1 - Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder - Google Patents

Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder Download PDF

Info

Publication number
US20150118306A1
US20150118306A1 US14/586,704 US201414586704A US2015118306A1 US 20150118306 A1 US20150118306 A1 US 20150118306A1 US 201414586704 A US201414586704 A US 201414586704A US 2015118306 A1 US2015118306 A1 US 2015118306A1
Authority
US
United States
Prior art keywords
sulforaphane
powder
enzyme
milk thistle
precursor
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
US14/586,704
Inventor
Brian Cornblatt
Grace Cornblatt
Anton Bzhelyansky
Robert Henderson
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.)
Nutramax Laboratories Inc
Original Assignee
Nutramax Laboratories 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=49882601&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150118306(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Nutramax Laboratories Inc filed Critical Nutramax Laboratories Inc
Priority to US14/586,704 priority Critical patent/US20150118306A1/en
Publication of US20150118306A1 publication Critical patent/US20150118306A1/en
Assigned to NUTRAMAX LABORATORIES, INC. reassignment NUTRAMAX LABORATORIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HENDERSON, ROBERT, BZHELYANSKY, Anton, CORNBLATT, Brian, CORNBLATT, Grace
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • 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
    • A23L33/105Plant extracts, their artificial duplicates or their derivatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • 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
    • 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
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/26Cyanate or isocyanate esters; Thiocyanate or isothiocyanate esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • 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
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • 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
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • 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
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/28Asteraceae or Compositae (Aster or Sunflower family), e.g. chamomile, feverfew, yarrow or echinacea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/31Brassicaceae or Cruciferae (Mustard family), e.g. broccoli, cabbage or kohlrabi
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01147Thioglucosidase (3.2.1.147), i.e. myrosinase
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs

Abstract

The invention relates to the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and a milk thistle extract or powder. The invention also relates to the combination of a sulforaphane or a derivative thereof and a milk thistle extract or powder. The invention also relates to the combination of a broccoli extract or powder and a milk thistle extract or powder. The invention provides compositions and methods relating to these combinations.

Description

  • This application claims priority to the following applications, each of which is incorporated by reference in its entirety: U.S. Provisional Patent Application No. 61/668,328, filed on Jul. 5, 2012; U.S. Provisional Patent Application No. 61/668,342, filed on Jul. 5, 2012; U.S. Provisional Patent Application No. 61/668,386, filed on Jul. 5, 2012; U.S. Provisional Patent Application No. 61/668,396, filed on Jul. 5, 2012; U.S. Provisional Patent Application No. 61/668,364, filed on Jul. 5, 2012; U.S. Provisional Patent Application No. 61,668,374, filed on Jul. 5, 2012; and U.S. Provisional Patent Application No. 61/794,417, filed on Mar. 15, 2013.
  • FIELD OF THE INVENTION
  • The present invention relates to the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and a milk thistle extract or powder. The present invention also relates to the combination of a sulforaphane or a derivative thereof and a milk thistle extract or powder. The present invention also relates to the combination of a broccoli extract or powder and a milk thistle extract or powder. The present invention provides compositions and methods relating to these combinations.
  • BACKGROUND OF THE INVENTION
  • The use of natural products is becoming increasingly popular with humans and companion animals. Some of these natural products are being incorporated into dietary supplements and medical foods. There is a need in the art for supplements which are useful as chemoprotective and/or antioxidant agents. In addition, there is a need in the art for pharmaceutical compositions and dietary supplements which are useful for conditions and disorders associated with glutathione. Chemoprotection through the use of natural products is evolving as a safe, effective, inexpensive, easily accessible, and practical means to prevent or reduce the occurrence of many conditions affecting humans and domesticated animals. It is known that carcinogens which can damage cells at the molecular level are often ingested and inhaled as non-toxic precursors. These non-toxic precursors may then convert into carcinogenic substances in the body. Chemoprotective agents, such as natural substances which can activate detoxifying enzymes or their co-factors, can counteract and allow for the elimination of carcinogens. These same natural substances can potentiate other naturally existing defenses such as the immune system.
  • Some natural products have antioxidant activity. Oxidative stress plays a major role in aging, the progression of neurodegenerative diseases as well as physiological trauma, such as ischemia. Antioxidant agents can reduce or inhibit the oxidation of vital biomolecules and may play a role in treating, preventing, or reducing the occurrence of conditions affected by oxidative stress.
  • Some natural products are useful for enhancing liver health. Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the U.S., affecting about 30% of the population. NAFLD is also known as hepatic lipidosis. In 10% of those with NAFLD, the disease will progress to non-alcoholic steatohepatitis (NASH), of which 25% will develop cirrhosis. Potentially 10-25% of those patients with cirrhosis will develop hepatocellular carcinoma. At the current rate and without effective treatment modalities, by 2030 hepatocellular carcinoma is projected to be the number one diagnosed cancer in the United States. NAFLD occurs when fat is deposited in the liver (steatosis), but not as a result of excessive alcohol use. NAFLD is associated with chronic inflammation, insulin resistance, diabetes and obesity. NAFLD presents few or no symptoms and is most commonly detected following abnormal results obtained during routine blood tests (i.e., elevated serum ALT and AST levels) and confirmed by an ultrasound or a biopsy. There are currently no surgical or pharmacological treatments for NAFLD. Recommendations to control NAFLD include lifestyle modifications such as eating a healthy diet, exercise, weight loss, lowering cholesterol and controlling diabetes.
  • NAFLD is often associated with a decrease in glutathione levels. Glutathione is a tripeptide with a gamma peptide linkage between the amine group of cysteine and the carboxyl group of the glutamate side chain. Glutathione plays an important role in the body, as it can serve as an antioxidant, detoxifier, and immunity enhancer. Glutathione can conjugate to metabolites and toxins, such as procarcinogens, for excretion from the body. Glutathione levels can be reduced in patients for a number of reasons, including poor diet, pollution, exposure to toxins and/or certain medications, stress, trauma, aging, infections, and radiation. Low levels of glutathione can cause a patient to be susceptible to oxidative stress, illness, and cancer. For example, reduced levels of glutathione are associated with conditions relating to the liver, prostate, brain, lung, kidneys, colon, breast, esophagus, pancreas, ovaries, etc. Examples of disorders associated with reduced levels of glutathione and glutathione deficiency include, but are not limited to: NAFLD, cancer (lung, prostate, colon, breast, brain, liver, ovarian, esophageal, pancreatic, nasopharyngeal, osteosarcoma), leukemia, cystic fibrosis, HIV, glutathione synthetase deficiency, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, multiple sclerosis, fibromyalgia, chronic fatigue, autism, and diabetes.
  • An example of a natural product thought to have chemoprotective and antioxidant properties is sulforaphane. Sulforaphane is an organosulfur compound which is also known as 1-isothiocyanato-4-methylsulfinylbutane. The sulforaphane precursor, glucoraphanin, can be obtained from vegetables of the Brassicaceae family, such as broccoli, brussels sprout, and cabbage. However, copious amounts of vegetables must be consumed in order to obtain levels adequate for chemoprevention. Glucoraphanin is converted into sulforaphane by a thioglucosidase enzyme called myrosinase, which occurs in a variety of exogenous sources such as Brassicaceae vegetables and endogenously in the gut microflora. However, upon ingestion of glucoraphanin, not all animals are capable of achieving its conversion to sulforaphane, most likely due to variations in microflora populations and overall health. In addition, in acidic environments such as the stomach, glucoraphanin can be converted to inert metabolites. The active metabolite, sulforaphane is able to induce nuclear factor erythroid-2-related factor (Nrf2) which, in turn, upregulates the production of Phase II detoxification enzymes and cytoprotective enzymes such as glutathione S-transferases, NAD(P)H:quinine oxidoreductase (NQO1), and heme-oxygenase-1 (HO-1). Sulforaphane has been thought to induce the production of these enzymes without significantly changing the synthesis of P-450 cytochrome enzymes. The upregulation of Phase II enzymes is thought to play a role in a variety of biological activities, including the protection of the brain from cytotoxicity, the protection of the liver from the toxic effects of fat accumulation, and the detoxification of a variety of other tissues.
  • Sulforaphane and its precursor glucoraphanin have been studied extensively. Shapiro et al. (Nutrition and Cancer, (2006), Vol. 55(1), pp. 53-62) discusses a clinical Phase I study determining the safety, tolerability, and metabolism of broccoli sprout glucosinolates and isothiocyanates. Shapiro et al. discusses a placebo-controlled, double-blind, randomized clinical study of sprout extracts containing either glucosinolates such as glucoraphanin or isothiocyanates such as sulforaphane in healthy human subjects. The study found that administration of these substances did not result in systematic, clinically significant, adverse effects.
  • Milk thistle (Silybum marianum) is a plant of the Asteraceae family. Milk thistle contains silymarin, which is composed of a number of constituents, including, but not limited to flavonolignans such as silibinin (also known as silybin or silibin), isosilibinin, silichristin, silydianin, kvercetin, dehydrosilybin, deoxysilycistin, deoxysilydianin, silandrin, silybinome, silyhermin and neosilyhermin. Silymarin constituents can have a number of biological effects, including inhibition of free radical formation, binding of free radical species, prevention of membrane lipid peroxidation, increase in levels of glutathione, and chelation of iron. Silibinin is the major active constituent of silymarin, and it thought that have hepatoprotective properties. Silymarin is discussed in U.S. Pat. No. 7,563,779, which is incorporated herein in its entirety.
  • Zhang et al. (Proc. Natl. Acad. Sci., (1994), Vol. 91, pp. 3147-3150) discusses a study in Sprague-Dawley rats to determine the anticarcinogenic activities of sulforaphane and structurally related synthetic norbornyl isiothiocyanates. The study determined that administration of sulforaphane was effective in blocking the formation of mammary tumors.
  • Cornblatt et al. (Carcinogenesis, (2007), Vol. 38(7): pp. 1485-1490) discusses a study in Sprague-Dawley rats to determine the effect of sulforaphane in chemoprevention in the breast. The study determined that oral administration of either sulforaphane or glucoraphanin resulted in a 3-fold increase in NAD(P)H:quinine oxidoreductase (NQO1) enzymatic activity and a 4-fold elevated immunostaining of the heme oxygenase-1 (HO-1) enzyme in the mammary epithelium.
  • Munday et al. (Cancer Res, (2008), Vol. 68(5): pp. 1593-1600) discusses a study regarding the effects of a freeze-dried aqueous extract of broccoli sprouts on bladder cancer development in rats. The study found that administration of the broccoli sprout extract resulted in a significant induction of glutathione S-transferase and NAD(P)H:quinine oxidoreductase 1 in the bladder, which are enzymes exerting protective activity against oxidants and carcinogens.
  • Aghazadeh S et al. (Exp Toxicol Pathol. (2011) September; 63(6):569-74) discuss the anti-apoptotic and anti-inflammatory effects of Silybum marianum in treatment of experimental steatohepatitis. The study found that administration of an extract of Silybum maranium to rats fed a methionine and choline deficient (MCD) diet to induce non-alcoholic steatohepatitis had improved AST and ALT activity along with an increase in the glutathione content compared to control rats being fed the MCD diet alone.
  • European Patent Application No. 2 213 280 discloses formulations comprising glucosinolates such as glucoraphanin, and myrosinase, wherein the formulation is encapsulated or coated.
  • All references cited herein are incorporated by reference in their entirety.
  • SUMMARY OF THE INVENTION
  • The present invention provides a composition comprising: (i) a sulforaphane precursor, preferably glucoraphanin; (ii) an enzyme capable of converting the sulforaphane precursor to sulforaphane, preferably a glucosidase enzyme, more preferably a thioglucosidase enzyme, and most preferably myrosinase; (iii) an enzyme potentiator, preferably ascorbic acid; and (iv) a milk thistle extract or powder. The present invention also provides a method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of, a disease or condition associated with the liver, prostate, brain, lung, kidneys, colon, breast, esophagus, pancreas, or ovaries in a subject, comprising administering to the subject: (i) a sulforaphane precursor, (ii) an enzyme capable of converting the sulforaphane precursor to sulforaphane, (iii) an enzyme potentiator, and (iv) a milk thistle extract or powder. The present invention also provides a method of increasing glutathione levels in a subject in need thereof in a subject, comprising administering to the subject: (i) a sulforaphane precursor, (ii) an enzyme capable of converting the sulforaphane precursor to sulforaphane, (iii) an enzyme potentiator, and (iv) a milk thistle extract or powder. The present invention also provides a method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a non-alcoholic fatty liver disease (NAFLD) and/or any other disorder of the liver, comprising administering to the subject: (i) a sulforaphane precursor, (ii) an enzyme capable of converting the sulforaphane precursor to sulforaphane, (iii) an enzyme potentiator, and (iv) a milk thistle extract or powder.
  • The present invention provides a composition comprising: (i) sulforaphane or a derivative thereof, and (ii) a milk thistle extract or powder. The present invention also provides a method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of, a disease or condition associated with the liver, prostate, brain, lung, kidneys, colon, breast, esophagus, pancreas, or ovaries in a subject, comprising administering to the subject: (i) sulforaphane or a derivative thereof, and (ii) a milk thistle extract or powder. The present invention also provides a method of increasing glutathione levels in a subject in need thereof in a subject, comprising administering to the subject, comprising administering to the subject: (i) sulforaphane or a derivative thereof, and (ii) a milk thistle extract or powder. The present invention also provides a method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a non-alcoholic fatty liver disease (NAFLD) and/or any other disorder of the liver, comprising administering to the subject: (i) sulforaphane or a derivative thereof, and (ii) a milk thistle extract or powder.
  • The present invention provides a composition comprising: (i) a broccoli extract or powder, and (ii) milk thistle extract or powder. The present invention also provides a method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of, a disease or condition associated with the liver, prostate, brain, lung, kidneys, colon, breast, esophagus, pancreas, or ovaries in a subject, comprising administering to the subject: (i) a broccoli extract or powder, and (ii) a milk thistle extract or powder. The present invention also provides a method of increasing glutathione levels in a subject in need thereof in a subject, comprising administering to the subject, comprising administering to the subject: (i) a broccoli extract or powder, and (ii) a milk thistle extract or powder. The present invention also provides a method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a non-alcoholic fatty liver disease (NAFLD) and/or any other disorder of the liver, comprising administering to the subject: (i) a broccoli extract or powder, and (ii) a milk thistle extract or powder.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a graph showing the conversion of glucoraphanin at 38° C. without ascorbic acid, as described in Example 4.
  • FIG. 2 is a graph showing the conversion within about 10 minutes at 38° C. as a function of ascorbic acid concentration, as described in Example 4.
  • FIG. 3 is a graph showing the conversion to sulforaphane within 30 minutes at 38° C. and 1 mM ascorbic acid, as described in Example 4.
  • FIG. 4 is a graph showing the conversion of glucoraphanin to sulforaphane in simulated intestinal fluid, as described in Example 5.
  • FIG. 5 is a graph showing the results of the experiment described in Example 6.
  • FIG. 6 is a graph showing the results of the experiment described in Example 6.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and a milk thistle extract or powder. The present invention also relates to the combination of sulforaphane or a derivative thereof and a milk thistle extract or powder. The present invention also relates to the combination of a broccoli extract or powder and a milk thistle extract or powder. The present invention also relates to the use of milk thistle extract or powder, with a mixture of one or more of the following: sulforaphane precursor, sulforaphane or a derivative thereof, and broccoli extract. The present invention provides compositions relating to these combinations.
  • The present invention provides methods comprising administering these combinations. In some embodiments, the combination may be administered for treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of, a disease or condition associated with the liver, prostate, brain, lung, kidneys, colon, breast, esophagus, pancreas, or ovaries in a subject, comprising administering to the subject. In some embodiments, the combination may be administered for increasing glutathione levels in a subject in need thereof in a subject. In some embodiments, the combination may be administered for treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a non-alcoholic fatty liver disease (NAFLD) and/or any other disorder of the liver in a subject.
  • Sulforaphane is also known as 1-isothiocyanato-4-methylsulfinylbutane. Derivatives of sulforaphane include, but are not limited to sulfoxythiocarbamate analogues of sulforaphane, 6-methylsulfinylhexyl isothiocyanate (6-HITC), and compounds which comprise the structure of sulforaphane with different side chains and/or various lengths of spacers between the isothiocyanato and sulfoxide groups. Examples of derivatives of sulforaphane include those described in the following references, each of which is incorporated herein by reference: Hu et al., Eur J Med Chem, 2013, 64:529-539; Ahn et al., Proc Natl Acad Sci USA, 2010, 107(21):9590-9595; and Morimistu et al., J. Biol. Chem. 2002, 277:3456-3463, and Baird et al., Arch Toxicol, 2011, 85(4):241-272.
  • In some embodiments, the composition comprises sulforaphane or a derivative thereof, preferably sulforaphane, in an amount of about 1 μg to about 10 g, preferably about 3 μg to about 5 g, preferably about 5 μg to about 1000 mg, preferably about 7 μg to about 750 mg, more preferably about 10 μg to about 500 mg, and most preferably about 100 μg to about 100 mg. In some embodiments, compositions suitable for human use comprise about 1 mg to about 20 mg.
  • In some embodiments, the methods of the present invention comprise administration of sulforaphane or a derivative thereof to a subject, preferably sulforaphane, in an amount of about 1 μg to about 10 g, preferably about 3 μg to about 5 g, preferably about 5 μg to about 1000 mg, preferably about 7 μg to about 750 mg, more preferably about 10 μg to about 500 mg, and most preferably about 100 μg to about 100 mg. In some embodiments wherein the subject is a human, the method comprises administration of about 1 mg to about 20 mg. In some embodiments, the methods of the present invention comprise administration of sulforaphane or a derivative thereof to a subject, preferably sulforaphane, in an amount of about 0.01 μg/kg to about 0.2 g/kg, preferably about 0.05 μg/kg to about 0.07 g/kg, more preferably about 0.07 μg/kg to about 15 mg/kg, more preferably about 0.1 μg/kg to about 11 mg/kg, and most preferably about 0.2 μg/kg to about 7 mg/kg. In some preferred embodiments wherein the subject is a human, the method comprises administration of about 2 μg/kg to about 2 mg/kg, and more preferably about 0.01 mg/kg to about 0.3 mg/kg. The above amounts may refer to each dosage administration or a total daily dosage. The total daily dosage refers to the total amount of a compound or ingredient which is administered to a subject in a twenty-four hour period.
  • In some embodiments, the method comprises administration of more than one of a sulforaphane or a derivative thereof. In some embodiments, the compositions comprise more than one of a sulforaphane or a derivative thereof. For example, the methods or composition may comprise both sulforaphane and one or more derivatives thereof, or two or more derivatives. In some embodiments wherein the method or composition comprise more than one of a sulforaphane or a derivative thereof, the above amounts may refer to the amount of each sulforaphane or a derivative thereof, or the total amount of the more than one sulforaphane or derivative thereof.
  • The term “sulforaphane precursor” refers to any compound, substance or material which can be used to produce sulforaphane. In preferred embodiments, the sulforaphane precursor comprises a compound which can be converted or metabolized to sulforaphane, preferably by an enzyme. In some preferred embodiments, the sulforaphane precursor comprises glucoraphanin. Glucoraphanin is a glucosinolate which is also known as 4-methylsulfinylbutyl glucosinolate and 1-S-[(1E)-5-(methylsulfinyl)-N-(sulfonatooxy)pentanimidoyl]-1-thio-β-D-glucopyranose.
  • In some embodiments, the composition comprises about 1 μg to about 10 g, preferably about 250 μg to about 5 g, more preferably about 500 μg to about 2000 mg, even more preferably about 1 mg to about 750 mg, even more preferably about 1.5 mg to about 250 mg, even more preferably about 2 mg to about 100 mg, and most preferably about 3 mg to about 75 mg of the sulforaphane precursor, preferably glucoraphanin. In some embodiments, compositions suitable for human use comprise about 3.5 mg to about 50 mg of the sulforaphane precursor, preferably glucoraphanin.
  • In some embodiments, the method comprises administering the sulforaphane precursor, preferably glucoraphanin to a subject, in an amount of about 1 μg to about 10 g, preferably about 250 μg to about 5 g, more preferably about 500 μg to about 2000 mg, even more preferably about 1 mg to about 750 mg, even more preferably about 1.5 mg to about 250 mg, even more preferably about 2 mg to about 100 mg, and most preferably about 3 mg to about 75 mg. In some embodiments wherein the subject is a human, the method comprises administration of about 3.5 mg to about 50 mg. In some embodiments, the method comprises administering an amount of sulforaphane precursor to a subject in an amount of about 1 μg/kg to about 1000 mg/kg, preferably about 5 μg/kg to about 500 mg/kg, more preferably about 7.5 μg/kg to about 100 mg/kg, even more preferably about 10 μg/kg to about 25 mg/kg, and most preferably about 25 μg/kg to about 10 mg/kg. In some embodiments wherein the subject is a human, the method comprises administration of about 50 μg/kg to about 800 μg/kg. The above amounts may refer to each dosage administration or a total daily dosage.
  • In some embodiments, the method comprises administration of more than one sulforaphane precursor. In some embodiments, the composition comprises more than sulforaphane precursor. In some embodiments wherein the method or composition comprises more than one sulforaphane precursor, the above amounts may refer to the amount of each sulforaphane precursor, or the total amount of the sulforaphane precursors.
  • The sulforaphane precursor may be converted or metabolized to sulforaphane. In some embodiments, the sulforphane precursor is converted to sulforaphane by an enzyme. In some embodiments, the enzyme capable of converting the sulforaphane precursor to sulforaphane comprises a glucosidase enzyme, preferably a thioglucosidase enzyme, and more preferably myrosinase. Myrosinase is also known as thioglucoside glucohydrolase.
  • In some embodiments, the composition comprises the enzyme in an amount of about 1 pg to about 1 ug, preferably about 50 pg to about 500 ng, and most preferably about 1 ng to about 150 ng. In some embodiments, compositions suitable for human use comprise about 5 ng to about 75 ng of the enzyme.
  • In some embodiments, the method comprises administering the enzyme, preferably myrosinase, in an amount of about 1 pg to about 1 μg, preferably about 50 pg to about 500 ng, and most preferably about 1 ng to about 150 ng. In some embodiments wherein the subject is a human, the method comprises administration of about 5 ng to about 75 ng of the enzyme. In some embodiments, the method comprises administering the enzyme to a subject in an amount of about 0.02 pg/kg to about 0.02 ug/kg, preferably about 0.7 pg/kg to about 7 ng/kg, and most preferably about 0.02 ng/kg to about 2 ng/kg. In some preferred embodiments wherein the subject is a human, the method comprises administration of about 0.1 ng/kg to about 1 ng/kg. The above amounts may refer to each dosage administration or a total daily dosage.
  • In some embodiments, the method comprises administration of more than one enzyme capable of converting the sulforaphane precursor to sulforaphane. In some embodiments, the composition comprises more than one enzyme capable of converting the sulforaphane precursor to sulforaphane. In some embodiments wherein the methods or compositions comprise more than one enzyme, the above amounts may refer to the amount of each enzyme, or the total amount of the enzymes.
  • The present invention also provides for the use of a broccoli extract and/or powder, including but not limited to broccoli seed and sprout extracts and powders. The present invention provides methods of administration of broccoli extract and/or powder, and compositions comprising broccoli extract and/or powder. In some embodiments, the broccoli extract or powder is standardized to contain about 1% to about 75% w/w, more preferably about 2.5% to about 50%, even more preferably about 5% to about 25%, and most preferably about 10% to about 20% of a sulforaphane precursor, preferably glucoraphanin. Examples of broccoli extracts and powders include but are not limited to those described in U.S. Pat. Nos. 5,411,986; 5,725,895; 5,968,505; 5,968,567; 6,177,122; 6,242,018; 6,521,818; 7,303,770, and 8,124,135, each of which is incorporated by reference in its entirety. Powders of broccoli may be obtained, for example, by air drying, freeze drying, drum drying, spray drying, heat drying and/or partial vacuum drying broccoli, preferably broccoli sprouts. In some embodiments, the compositions and methods comprise use of about 1 μg to about 10 g, more preferably about 250 μg to about 5 g, even more preferably about 500 μg to about 1 g, preferably about 600 μg to about 500 mg, more preferably about 750 μg to about 400 mg, and most preferably about 1 mg to about 300 mg of the broccoli extract. In some embodiments, the broccoli extract or powder is present in a composition or administered to a subject in amounts sufficient to provide a sulforaphane precursor or sulforaphane in the amounts described above. In some embodiments, the composition may further comprise an enzyme potentiator, preferably ascorbic acid. In some embodiments, the method may further comprise administration of an enzyme potentiator, preferably ascorbic acid.
  • The sulforaphane or a derivative thereof, the sulforaphane precursor, and/or the enzyme capable of converting the sulforaphane precursor to sulforaphane may be obtained from any source, including but not limited to one or more plants from the Brassicaceae (also known as Cruciferae) family. Examples of plants from the Brassicaceae family include, but are not limited to, the following: broccoli, Brussels sprouts, cauliflower, cabbage, horseradish, parsnip, radish, wasabi, watercress, and white mustard. In some preferred embodiments, sulforaphane precursor, preferably glucoraphanin, and the enzyme, preferably myrosinase, are obtained from broccoli, broccoli sprouts, or broccoli seeds. The sulforaphane precursor and the enzyme may be obtained from the same source or from different sources. In some embodiments, both the sulforaphane precursor and the enzyme may be obtained from an extract or powder from these plants, preferably a broccoli seed or sprout extract or powder.
  • The present invention provides for the use of an enzyme potentiator. Enzyme potentiators may be used to enhance the activity of the enzyme that is capable of converting the sulforaphane precursor to sulforaphane. In some embodiments, the enzyme potentiator comprises an enzyme co-factor, preferably ascorbic acid. Ascorbic acid, also known as ascorbate or vitamin C, can potentiate the activity of myrosinase. In some embodiments, without an enzyme potentiator such as ascorbic acid, the conversion reaction to sulforaphane may be too slow to occur in the location needed for peak absorption. The enzyme potentiator may be obtained from a natural source, or it may be produced synthetically.
  • In some embodiments, the compositions may comprise about 1 mg to about 500 mg, preferably about 1 mg to about 250 mg, and most preferably about 1 mg to about 125 mg of the enzyme potentiator. In some preferred embodiments, compositions suitable for human use comprise about 1 mg to about 50 mg of the enzyme potentiator.
  • In some embodiments, the method of the present invention comprises administration of an enzyme potentiator, preferably ascorbic acid, in an amount of about 1 mg to about 500 mg, preferably about 1 mg to about 250 mg, and most preferably about 1 mg to about 125 mg. In some preferred embodiments wherein the subject is a human, the method comprises administration of about 1 mg to about 50 mg. In some embodiments, the method of the present invention comprises administration of the enzyme potentiator, preferably ascorbic acid, in an amount of about 0.01 mg/kg to about 3 mg/kg, and most about 0.02 mg/kg to about 2 mg/kg. In some preferred embodiments wherein the subject is a human, the method comprises administration of about 0.02 mg/kg to 0.7 mg/kg of the enzyme potentiator. The above amounts may refer to each dosage administration or a total daily dosage.
  • In some embodiments, the method comprises administration of more than one enzyme potentiator. In some embodiments, the composition comprises more than one an enzyme potentiator. In some embodiments wherein the method or composition comprise more than one enzyme potentiator, the above amounts may refer to the amount of each enzyme potentiator, or the total amount of the enzyme potentiators.
  • The present invention further comprises the use of a milk thistle extract or powder. Milk thistle belongs to the species Silybum marianum. Milk thistle comprises a number of components or fractions having biological activity. An active fraction of milk thistle is silymarin, which comprises a number of constituents. Examples of constituents of silymarin include, but are not limited to: silibinin (also known as silybin or silibin), isosilibinin, silichristin, silydianin, and kvercetin, dehydrosilybin, deoxysilycistin, deoxysilydianin, silandrin, silybinome, silyhermin and neosilyhermin. Isomers of silibinin include Silybinin A (or Silibinin A) and Silybinin B (or Silbinin B). In preferred embodiments, the milk thistle extract or powder comprises silymarin.
  • In some embodiments, the compositions and methods of the present invention may comprise the use of one or more derivatives of silymarin, instead of or in addition to a milk thistle extract or powder. Derivatives of silymarin constituents include any modified forms of the above compounds, including but not limited to, 7-0- and 23-O-acyl derivatives, and analogues. Examples of derivatives of silymarin constituents include, but are not limited to 2,3-dehydrosilybin (DHS); 7-O-methylsilybin; 7-O-galloylsilybin; 7,23-disulphatesilybin (DSS); 7-O-palmitoylsilybin; and 23-O-palmitoylsilybin. Examples of derivatives include those described in the following references, each of which is incorporated herein by reference in its entirety: Agarwal et al. PLOS ONE, 2013, 8(3):e60074; GB 2167414; and CA1337124. In some embodiments, the compositions and methods of the present invention may comprise the use of silymarin or silibinin in a purified form or silymarin or silibinin produced synthetically, instead of or in addition to a milk thistle extract or powder.
  • In some embodiments, the milk thistle extract or powder may be used. In preferred embodiments, the milk thistle extract comprises silymarin. In preferred embodiments, the milk thistle extract comprises silibinin. In some embodiments, the milk thistle extract or powder is standardized to contain about 25% to about 95%, preferably about 50% to about 90%, and more preferably about 55% to about 85% silymarin. In some embodiments, the milk thistle extract or powder is standardized to contain about 5% to about 75%, preferably about 10% to about 60%, more preferably about 15% to about 50%, and most preferably about 20% to about 35% of silibinin. Examples of milk thistle extract include, but are not limited to, those described in U.S. Pat. No. 6,555,141; U.S. Pat. No. 6,863,906; U.S. Pat. No. 7,563,779; WO200908006; EP2020238; WO2009043671; EP1584240; and WO2011076154, each of which is incorporated by reference in its entirety. Powders of milk thistle may be obtained, for example, by air drying, freeze drying, drum drying, spray drying, heat drying and/or partial vacuum drying milk thistle.
  • In some embodiments, the compositions and methods comprise use of about 1.25 mg to about 15 grams, preferably about 5 mg to about 10 grams, and most preferably about 10 mg to about 7.5 grams of milk thistle extract. In some preferred embodiments wherein the composition is suitable for human use, the composition comprises about 25 mg to about 5 grams of the milk thistle extract. In some embodiments, the composition comprises about 0.75 mg to about 15 grams, preferably about 3 mg to about 7 grams, more preferably about 7 mg to about 5 grams, and most preferably about 15 mg to about 3.5 grams of silymarin. In some preferred embodiments wherein the composition is suitable for human use, the composition comprises about 50 mg to about 200 mg of silymarin. In some embodiments, the composition comprises about 0.3 mg to about 5 grams, preferably about 1.5 mg to about 3 grams, more preferably about 3 mg to about 2 grams, and most preferably about 7 mg to about 1.5 grams of silibinin. In some preferred embodiments wherein the composition is suitable for human use, the composition comprises about 30 mg to about 90 mg of silibinin.
  • In some embodiments, the method comprises administration of silymarin in an amount of about 1 μg/kg to about 75 mg/kg, preferably about 2.5 μg/kg to about 50 mg/kg, more preferably about 5 μg/kg to about 25 mg/kg, more preferably about 10 μg/kg to about 15 mg/kg, and most preferably about 15 μg/kg to about 10 mg/kg. In some preferred embodiments wherein the subject is a human, the method comprises administration of about 1 mg/kg to about 3 mg/kg of silymarin. In some embodiments, the method comprises administration of silibinin in an amount of about 0.5 μg/kg to about 50 mg/kg, preferably about 1 μg/kg to about 35 mg/kg, more preferably about 2.5 μg/kg to about 25 mg/kg, more preferably about 5 μg/kg to about 10 mg/kg, and most preferably about 7.5 μg/kg to about 5 mg/kg. In some preferred embodiments wherein the subject is a human, the method further comprises administration of about 300 μg/kg to about 2 mg/kg of silibinin. In embodiments wherein a derivative of a silymarin constituent are used, the methods and compositions may comprise the derivative in an amount of about 0.75 mg to about 15 grams, preferably about 3 mg to about 7 grams, more preferably about 7 mg to about 5 grams, and most preferably about 15 mg to about 3.5 grams. In some preferred embodiments wherein the composition is suitable for human use, the compositions comprise about 50 mg to about 200 mg of the derivative of a silymarin constituent. The above amounts may refer to each dosage administration or a total daily dosage.
  • In some embodiments, S-adenosylmethionine may be used in place of, or in addition to, the milk thistle extract, silymarin and/or silibinin.
  • The methods of the present invention may further comprise administration of one or more additional components. The compositions of the present invention may further comprise one or more additional components. The additional components may include active pharmaceutical ingredients, nutritional supplements, and nutritional extracts. Examples of additional components include, but are not limited, quercetin or a derivative thereof, an aminosugar such as glucosamine, a glycosaminoglycan such as chondroitin, avocado/soybean unsaponifiables, vitamins such as vitamin K2, coffee fruit, magnesium, ursolic acid, proanthocyanidins, alpha- and beta-glucans, curcumin, phytosterols, phytostanols, and S-adenosylmethionine (SAMe). These additional components may be present in cranberry (Vaccinium macrocarpon) extract (proanthocyanidins, quercetin, and ursolic acid), turmeric (Curcuma longa), medicinal mushroom extract such as shiitake (Lentinus edodes), maitake (Grifola frondosa) mushroom extracts, and reishi (Ganoderma lucidum) mushroom extract.
  • In some embodiments, the ratio of silymarin to sulforaphane or a derivative of is about 1:1 to about 75:1, more preferably about 2:1 to about 50:1, more preferably about 2.5:1 to about 25:1, even more preferably about 5:1 to about 15:1, and most preferably about 6:1 to about 9:1. In some embodiments, the ratio of silibinin to sulforaphane or a derivative of is about 1:2 to about 35:1, more preferably about 1:1 to about 25:1, more preferably about 1:1 to about 15:1, even more preferably about 2:1 to about 10:1, and most preferably about 2:1 to about 5:1. In some embodiments, the ratio of silymarin to sulforaphane precursor of is about 1:5 to about 50:1, preferably about 1:2 to about 25:1, more preferably about 1:1 to about 10:1, more preferably about 1.5:1 to about 5:1, and most preferably about 1:1 to about 4:1. In some embodiments, the ratio of silibinin to sulforaphane precursor is about 1:5 to about 50:1, preferably about 1:2 to about 25:1, preferably 1:1 to about 20:1, and most preferably about 1:1 to about 13:1.
  • In some embodiments, the composition comprises a unit dosage form, including but not limited to pharmaceutical dosage forms suitable for oral, rectal, intravenous, subcutaneous, intramuscular, transdermal, transmucosal, and topical. In some preferred embodiments, the composition comprises an orally administrable dosage form or a rectally administrable dosage form. Examples of orally administrable dosage forms include, but are not limited to a tablet, capsule, powder that can be dispersed in a beverage, a liquid such as a solution, suspension, or emulsion, a soft gel/chew capsule, a chewable bar, or other convenient dosage form known in the art. In preferred embodiments, the composition comprises a tablet, capsule, or soft chewable treat. The orally administrable dosage forms may be formulated for immediate release, extended release or delayed release.
  • In some embodiments, at least the sulforaphane precursor, the enzyme, and the enzyme potentiator are provided in a dosage form which allows for the release in an area of the gastrointestinal tract having a pH of at least 4 and preferably at least 5, such as the small intestine, preferably the duodenum. In some embodiments, at least the sulforaphane or derivative thereof and/or the broccoli extract or powder are provided in a dosage form which allows for the release in an area of the gastrointestinal tract having a pH of at least 4 and preferably at least 5, such as the small intestine, preferably the duodenum. In some embodiments, the milk thistle extract or powder and/or any optional additional components are also released in an area of the gastrointestinal tract having a pH of at least 4 and preferably at least 5, such as the small intestine, preferably the duodenum. The small intestine includes the duodenum, jejunum, and ileum.
  • In some embodiments, each of these components (i.e, sulforaphane precursor, enzyme, enzyme potentiator, sulforaphane or a derivative thereof, broccoli extract or powder, milk thistle extract or powder, and/or additional components) are released simultaneously or concomitantly (i.e., within a short period of time of each other). This provides benefits over glucoraphanin-containing compositions formulated to release the glucoraphanin in an area of the gastrointestinal tract having a pH below 4, such as the stomach. In low pH environments such as this, the acidic environment may divert conversion of sulforaphane precursor to other, physiologically inactive end products, such as sulforaphane nitrile and epithionitrile.
  • In some embodiments, the compositions may comprise orally administrable compositions which comprise gastroprotective formulations, including enteric coated dosage forms or any dosage form which is resistant to degradation in an area of the gastrointestinal tract having pH below 4, such as the stomach. For example, the orally administrable composition may comprise a tablet or capsule comprising an enteric coating. The enteric coating may comprise materials including, but not limited to cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, polyvinyl acetate phthalate, methacrylic acid copolymer, methacrylic acid:acrylic ester copolymer, hydroxypropyl methylcellulose acetate succinate, hydroxypropyl methylcellulose trimellitate, shellac, cellulose acetate trimellitate, carboxymethylethylcellulose, and mixtures thereof. The enteric coating may comprise any suitable enteric polymers known in the art. In some embodiments, one or more of the components in the composition may be embedded in a matrix of enteric polymers. In some embodiments, the orally administrable compositions comprise a capsule that dissolves slowly in gastric acid and travels to the small intestine, such as DRCAPS™ acid resistant capsules, which are marketed by CAPSUGEL® or any other acid resistant capsules.
  • In the most preferred form, the orally administrable composition is surrounded by a coating that does not dissolve unless the surrounding medium is at a pH of at least 4, and more preferably at least 5. Alternatively, a coating may be employed which controls the release by time, as opposed to pH, with the rate adjusted so that the components are not released until after the pH of the gastrointestinal tract has risen to at least 4, and more preferably at least 5. Thus, a time-release formulation may be used to prevent gastric presence of the sulforaphane precursor, the enzyme capable of converting the sulforaphane precursor to sulforaphane, and the enzyme potentiator, or of the sulforaphane. The coating layer(s) may be applied onto orally administrable composition using standard coating techniques. The enteric coating materials may be dissolved or dispersed in organic or aqueous solvents. The pH at which the enteric coat will dissolve can be controlled by a polymer, or combination of polymers, selected and/or ratio of pendant groups. For example, dissolution characteristics of the polymer film can be altered by the ratio of free carboxyl groups to ester groups. Enteric coating layers also contain pharmaceutically acceptable plasticizers such as triethyl citrate, dibutyl phthalate, triacetin, polyethylene glycols, polysorbates or other plasticizers. Additives such as dispersants, colorants, anti-adhering and anti-foaming agents may also be included.
  • The compositions may contain one or more non-active pharmaceutical ingredients (also known generally as “excipients”). Non-active ingredients, for example, serve to solubilize, suspend, thicken, dilute, emulsify, stabilize, preserve, protect, color, flavor, and fashion the active ingredients into an applicable and efficacious preparation that is safe, convenient, and otherwise acceptable for use. The excipients are preferably pharmaceutically acceptable excipients. Examples of classes of pharmaceutically acceptable excipients include lubricants, buffering agents, stabilizers, blowing agents, pigments, coloring agents, flavoring agents, fillers, bulking agents, fragrances, release modifiers, adjuvants, plasticizers, flow accelerators, mold release agents, polyols, granulating agents, diluents, binders, buffers, absorbents, glidants, adhesives, anti-adherents, acidulants, softeners, resins, demulcents, solvents, surfactants, emulsifiers, elastomers and mixtures thereof.
  • In some embodiments, the combination of (i) a sulforaphane precursor, preferably glucoraphanin, (ii) an enzyme capable of converting the sulforaphane precursor to sulforaphane, preferably a glucosidase enzyme, more preferably a thioglucosidase enzyme, and most preferably myrosinase, (iii) an enzyme potentiator, preferably an enzyme co-factor, more preferably ascorbic acid, and (iv) milk thistle extract or powder demonstrates a synergistic effect. In some embodiments, the combination of sulforaphane (or a derivative thereof) and a milk thistle extract or powder demonstrates a synergistic effect. Synergy refers to the effect wherein a combination of two or more components provides a result which is greater than the sum of the effects produced by the agents when used alone. In preferred embodiments, the synergistic effect is greater than an additive effect. In some embodiments, the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and a milk thistle extract or powder has a statistically significant, greater effect compared to: (i) each component alone, (ii) the combination of sulforaphane precursor and the enzyme alone; and/or (iii) the combination of sulforaphane precursor, the enzyme, and the enzyme potentiator alone.
  • In preferred embodiments, the combination of the sulforaphane precursor, the enzyme, the enzyme potentiator, and a milk thistle extract or powder demonstrates synergy by having a statistically significant and/or greater than additive effect compared to the sulforaphane precursor alone and the milk thistle extract or powder alone. In some embodiments, the combination of glucoraphanin, myrosinase, ascorbic acid, and silymarin has a synergistic effect compared to the combination of glucoraphanin, myrosinase, ascorbic acid alone; and compared to silymarin alone. In some embodiments, the combination of glucoraphanin, myrosinase, ascorbic acid, and silibinin has a synergistic effect compared to the combination of glucoraphanin, myrosinase, ascorbic acid alone; and compared to silibinin alone.
  • In some embodiments, the combination of a sulforaphane (or a derivative thereof) and a milk thistle extract or powder has a statistically significant and/or greater than additive effect than: (i) sulforaphane (or a derivative thereof) alone, and/or (ii) a milk thistle extract or powder alone. In some embodiments, the combination of sulforaphane and silymarin has a synergistic effect compared to sulforaphane alone, and silymarin alone. In some embodiments, the combination of sulforaphane and silibinin has a synergistic effect compared to sulforaphane alone, and silibinin alone.
  • In some embodiments, the combination of broccoli extract or powder and a milk thistle extract or powder has a statistically significant and/or greater than additive effect than: (i) broccoli extract or powder alone, and/or (ii) a milk thistle alone. In some embodiments, the combination of broccoli extract or powder and silymarin has a synergistic effect compared to broccoli extract or powder alone, and silymarin alone. In some embodiments, the combination of broccoli extract or powder and silibinin has a synergistic effect compared to broccoli extract or powder alone, and silibinin alone.
  • The present invention provides methods of use, including methods of administration to a subject in need thereof. In some embodiments, the method comprises administration of the combination of a sulforaphane precursor, an enzyme capable of converting the sulforaphane precursor to sulforaphane, an enzyme potentiator, and a milk thistle extract or powder. In some embodiments, the method comprises administration of the combination of a sulforaphane or a derivative thereof and a milk thistle extract or powder. In some embodiments, the method comprises administration of the combination of a broccoli extract or powder and a milk thistle extract or powder.
  • In some embodiments, the method relates to treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of, a disease or condition associated with the liver, genitourinary system (including prostate, breast, and ovaries), brain, lung, kidneys, colon, esophagus, pancreas, or hematopoietic system in a subject, comprising administering to the subject. The methods may be useful in reducing damage of slowing damage to tissues and organs, such as the liver, genitourinary system (including prostate, breast, and ovaries), brain, lung, kidneys, colon, esophagus, and pancreas, In some embodiments, the method relates to increasing glutathione levels in a subject in need thereof in a subject. The method may also be useful in treating, preventing, decreasing the symptoms associated with, and/or reducing secondary recurrences of diseases or conditions associated with low levels of glutathione. Examples of such diseases and conditions include, but are not limited to, non-alcoholic fatty liver disease (NAFLD), cancer (such as cancer of the liver, lung, prostate, colon, breast, brain, ovaries, esophagus, pancreas, nasopharynx, osteosarcoma), leukemia, cystic fibrosis, HIV, glutathione synthetase deficiency, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, multiple sclerosis, fibromyalgia, chronic fatigue, autism, diabetes, hepatotoxicity, and toxicity due to environmental factors.
  • In some embodiments, method relates to treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and/or reducing secondary recurrences of a non-alcoholic fatty liver disease (NAFLD) and/or any other disorder of the liver in a subject. In some embodiments, the methods relate to ameliorating or reducing the deleterious effects of NAFLD.
  • In some embodiments, the methods relate to providing a beneficial effect on biomarkers, and treating, preventing, reducing the occurrence of, decreasing the symptoms associated with abnormal levels of these biomarkers. Examples of such biomarkers include, but are not limited to NADPH-dependent enzymes, thioredoxin (TXN), thioredoxin reductase-1 (Txnrd-1), glutamate-cysteine ligase subunit (GCLC), sulfotransferase 1A1 (SULT1A1), heme oxygenase-1 (HMOX1), glutathione peroxidase-3 (GPx-3), glutathione S-transferse theta 2 (GSTT2), microsomal glutathione S-transferase 1 (MGST1), aldehyde oxidase (AOX1), aldo-keto reductase 1B8 (Akr1 b8), flavin-containing monooxygenase 2 (FMO2), Fc receptor region receptor III (Fcgr3), tryptase beta 1 (TPSB1), mast cell protease-6 (Mcpt6), neurexin-1-alpha (NRXN-1), microphthalmia-associated transcription factor (MITF), type II iodothyronine deiodinase (DIO2), angiopoietin-14 (Angpt14), cluster of differentiation (CD36), and Ntel. Diseases or conditions associated with elevated or abnormal levels of these biomarkers include, but are not limited to cancer, pulmonary and central nervous system tuberculosis, multiple sclerosis, Crohn's disease, atherosclerosis, osteoarthritis, asthma, stroke, emphysema, diabetic nephropathy, chronic histiocytic intervillositis of the placenta, hypertension, abdominal aortic aneurysm, inflammatory bowel disease, chronic rhinosinusitis, coronary artery disease, and kidney disease.
  • In some embodiments, the method comprises administering to a subject in need thereof a combination of sulforaphane and a milk thistle extract or powder. In some embodiments the method comprises administering to a subject in need thereof a combination of broccoli extract or powder and a milk thistle extract or powder. In some preferred embodiments, the method comprises administering to the subject a combination of glucoraphanin, myrosinase, ascorbic acid, and a milk thistle extract or powder. In preferred embodiments, the combinations demonstrate a synergistic effect in the methods of the present invention.
  • In preferred embodiments, one or more components of the combinations (for example, the sulforaphane precursor, the enzyme capable of converting the sulforaphane precursor to sulforaphane, the enzyme potentiator, the milk thistle extract or powder; or the sulforaphane or derivative thereof and the milk thistle extract or powder; or the broccoli extract or powder and the milk thistle extract or powder) are administered together in one composition or dosage form, or separately, preferably within a period in which their therapeutic properties overlap. In some embodiments, the components of the combinations may be administered in two or more orally administrable compositions or dosage forms. For example, in some embodiments, the sulforaphane precursor, the enzyme capable of converting the sulforaphane precursor to sulforaphane, and the enzyme potentiator are administered in one orally administrable dosage form, while the a milk thistle extract or powder are administered in one or more separate or additional orally administrable dosage form(s). In preferred embodiments, the components of the combination are administered in one dosage form.
  • In some embodiments, the combination may be administered at a frequency of 1 to 10 times daily, preferably 1 to 5 times daily, more preferably 1 to 3 times daily, and most preferably 1 time daily.
  • The dosages disclosed in this application refer generally to dosages suitable for humans. Dosage calculations can be determined by those of skilled in the art by evaluating body weight, surface area, metabolic rate, and species differences.
  • The term “subject” refers to any animal, including mammals and birds. Mammals include, but are not limited to, humans, dogs, cats, horses, cows, camels, elephants, lions, tigers, bears, seals, and rabbits. In preferred embodiments, the subjects comprise mammals that are not consumed as food, such as humans, cats, and dogs.
  • EXAMPLES Example 1
  • The following is an exemplary formulation:
  • Glucoraphanin-containing broccoli extract (about 12% w/w), 50 mg to 5 g
    Myrosinase-containing freeze-dried broccoli sprout powder, 25 mg to 500 mg
    Ascorbic acid, 5 mg to 500 mg
    Milk thistle extract (about 20% to 35% w/w silibinin), 25 mg to 5 g
  • Example 2
  • A Hydrophobic Interaction Chromatographic (HILIC) method was developed, comprising the following conditions:
  • Column: Waters BEH Amide, 1.7-μm particle size; 2.1 mm×100 mm
  • Mobile Phase: 20% 10 mM Ammonium Acetate, pH 5.0; 80% Acetonitrile;
  • Separation mode: isocratic
  • Column Temperature: 70° C.
  • Flow Rate: 0.7 mL/min
    The above conditions allow separation of five typical Brassicaceae glucosinolates, including the sulforaphane precursor, glucoraphanin.
  • Example 3 Consumption of Glucoraphanin as a Function of the Ascorbic Acid Concentration
  • About 250 mg of broccoli seed extract containing about 12% (w/w) glucoraphanin were subjected to hydrolysis by a fixed concentration of broccoli sprout-derived myrosinase in the presence of variable concentration of ascorbic acid, ranging from 0 to 600 μmoles/Liter. The reaction mixtures were thermostated at 38° C.; aliquots were withdrawn every 15 minutes for 60 minutes, and concentration of glucoraphanin determined chromatographically. The rate of glucoraphanin consumption was interpreted as the rate its conversion to sulforaphane. Graphical representation of glucoraphanin content reduction as a function of increasing ascorbic acid concentration results in a series of linear plots; the slopes of the linear regression lines reflect the rate of glucoraphanin consumption, in μmoles/minute. It is apparent that in the presence of 600 μmoles/Liter concentration of ascorbic acid, the reaction rate increased 13-fold relative to that which proceeded in the absence of modulatory effects of ascorbic acid.
  • Content of Ascorbic Acid
    250 μM
    Time, min 0 μM 50 μM 125 μM 250 μM Filtered 400 μM 600 μM
    0 93.36 93.36 93.36 93.36 93.36 93.36 93.36 μmoles
    15 92.24 89.20 84.52 80.95 86.31 78.32 75.02 GR
    30 90.71 84.24 75.92 69.06 79.44 62.78 55.66
    45 89.44 80.30 68.09 57.63 71.94 47.67 37.50
    60 87.79 76.36 59.41 45.76 65.18 33.15 22.09
    Slope −0.09293 −0.28599 −0.56217 −0.79012 −0.47140 −1.00714 −1.20029 μmol/min
    Intercept 93.496 93.271 93.123 93.053 93.386 93.270 92.734 μmol
  • Example 4 Equimolar Conversion of Glucoraphanin to Sulforaphane
  • A two-part experiment was conducted to further elucidate the role of ascorbic acid in modulating myrosinase activity. All solutions were prepared in 20 mM Tris-buffered saline, at pH 7.5, previously identified as an optimal for myrosinase activity; each sample tube had 100 mg of freeze-dried broccoli powder accurately weighed in as a source of myrosinase. Experiment was conducted at 38° C. for 2 hours, with sample aliquots removed in 30-minute increments, and both glucoraphanin and sulforaphane content assessed by HPLC. A strongly acidic “stop” solution was utilized to instantaneously inhibit further myrosinase activity in the removed aliquots. A control sample contained no ascorbic acid, and the enzymatic conversion proceeded unassisted by a co-factor.
  • PART 1. In the presence of the fixed concentration of ascorbic acid, 1 mmol/Liter, an increasing amount of broccoli seed extract (about 12% glucoraphanin, w/w) was added, ranging from 250 mg to 500 mg.
  • PART 2. While keeping the amount of broccoli seed extract fixed at 250 mg, the concentration of ascorbic acid was varied from 0.4 mmol/Liter to 3.8 mmol/Liter.
  • The table below presents glucoraphanin and sulforaphane expressed in μmoles. It is apparent that within the first 30 minutes in almost all the reaction mixtures, conversion of glucoraphanin to sulforaphane was complete. However, careful examination of the enzymatic conversion occurring in the control sample, without the stimulating effects of ascorbic acid, reveals an equimolar conversion of glucoraphanin to sulforaphane, i.e., the amount of glucoraphanin consumed results in the equivalent amount of sulforaphane produced.
  • Glucoraphanin, μmoles Sulforaphane, μmoles
    Time, min
    0 30 60 90 120 0 30 60 90 120
    GR 250 mg AA 0.0 mM 58.02 48.57 37.52 26.58 15.67 3.42 12.08 22.27 33.17 42.89
    GR 250 mg AA 1.0 mM 40.07 21.51 61.95 60.20 60.04 58.25
    GR 300 mg AA 1.0 mM 49.31 24.18 74.40 73.04 72.19 70.56
    GR 350 mg AA 1.0 mM 61.41 25.00 84.92 84.02 83.19 80.02
    GR 400 mg AA 1.0 mM 71.35 1.56 26.71 96.60 95.38 93.39 91.16
    GR 500 mg AA 1.0 mM 89.41 1.01 33.52 120.16 118.45 116.45 112.34
    GR 250 mg AA 0.4 mM 45.66 15.98 62.06 61.01 60.88 58.90
    GR 250 mg AA 1.0 mM 35.24 26.49 62.19 60.62 60.41 59.10
    GR 250 mg AA 2.0 mM 24.94 36.05 60.85 59.78 59.65 58.08
    GR 250 mg AA 2.9 mM 22.24 38.20 59.95 59.34 58.77 56.99
    GR 250 mg AA 3.8 mM 21.70 37.87 58.77 57.79 58.41 56.17
  • In the Part 2 of the experiment, the modulatory effect of the increasing concentration of ascorbic acid on the activity of myrosinase was assessed. An initial, apparently linear, increase in myrosinase-promoted conversion of glucoraphanin to sulforaphane is observed to about 2 mmol/L of ascorbic acid concentration, followed subsequently by a considerable leveling off.
  • Finally, examination of sulforaphane yield of after 30 minutes within the PART 1 of the experiment, reveals that in the presence of 1 mmol/Liter of ascorbic acid, the fixed amount of myrosinase contained in 100 mg of freeze-dried broccoli sprout powder is capable of generating at least 200 μmoles of sulforaphane, in a predictably linear fashion. FIGS. 1, 2, 3, and 4 demonstrate the results of this study.
  • Example 5 Conversion of Glucoraphanin to Sulforaphane in the Presence of Simulated Intestinal Fluid
  • Simulated Intestinal Fluid (SIF) powder, a commercially supplied concentrate closely approximating the human intestinal content in terms of composition, pH and ionic strength, was used. The experiment utilized a USP Dissolution Apparatus 2 (paddles), where into six dissolution vessels 500 mL of Simulated Intestinal Fluid was dispensed, along with 150 mg of freeze-dried broccoli sprout powder as a source of myrosinase. In vessels 1-4, the concentration of ascorbic acid was varied from 0.25 to 1.00 mmol/Liter; in vessel 5, in addition to 1 mmol/Liter ascorbic acid, 3.125 g of pancreatin (8×USP) was suspended; in vessel 6, in addition to 1 mmol/Liter ascorbic acid, and 3.125 g of pancreatin (8×USP), a doubled amount of freeze-dried broccoli sprout powder (300 mg) was added. After vessels were brought to 38° C., 250 mg of glucoraphanin-rich (12%, w/w) broccoli seed extract was added to each, and the resulting suspensions were stirred at 75 RPM for 2 hours. Aliquots were withdrawn every 15 minutes, and assayed for sulforaphane. FIG. 4 shows direct correlation between larger yield of sulforaphane and higher concentrations of ascorbic acid, especially at the earlier stages of the experiment.
  • Example 6
  • The following study was conducted to determine the effect of the combination of sulforaphane and silibinin on glutathione levels. Glutathione plays an important role in the body, as it can serve as an antioxidant, detoxifier, and immunity enhancer. Decreased levels of glutathione can cause a patient to be susceptible to oxidative stress, illness, and cancer. Therefore, an increase in glutathione levels is a beneficial effect.
  • In the study, the human liver cancer cell line, HepG2 cells were treated with DMSO (vehicle control), sulforaphane (SFN), silibinin (Silib), or the combination of sulforaphane and silibinin, for 24 hours. Cell lysates were collected and glutathione levels were measured using o-phthalaldehyde (OPT) as a fluorescent reagent. FIG. 5 and FIG. 6 show the results of the study.
  • In Part 1 of the study, the effect of 0.5 μM SFN was compared to various concentrations of silibinin and to the combination of 0.5 μM SFN and silibinin at various concentrations. In particular, the cells were treated with one of the following: (i) DMSO (vehicle control), (ii) 0.5 μM SFN, (iii) 100 μM Silib, (iv) 200 μM Silib, (v) 300 μM Silib, (vi) 0.5 μM SFN and 100 μM Silib, (vii) 0.5 μM SFN and 200 μM Silib, and (viii) 0.5 μM SFN and 300 μM Silib. The results demonstrate that the combination of sulforaphane and silibinin at each of the tested dosages had a synergistic effect compared to each component alone. For example, when the cells were treated with individual components the glutathione levels remained the same with treatment of sulforaphane alone or decreased slightly with Silibinin treatment compared the DMSO (vehicle) control. However, when cells were treated with the combination of sulforaphane and silibinin, at each of the tested dosages, the glutathione levels synergistically increased compared to the control. An increase in glutathione levels is a beneficial effect. The results are depicted in FIG. 5.
  • In Part 2 of the study, the effect of 2 μM SFN was compared to various concentrations of silibinin and to the combination of 2 μM SFN and silibinin at various concentrations. In particular, the cells were treated with one of the following: (i) DMSO (vehicle control), (ii) 2 μM SFN, (iii) 100 μM Silib, (iv) 300 μM Silib, (v) 2 μM SFN and 100 μM Silib, and (vi) 0.5 μM SFN and 300 μM Silib. The results demonstrate that the combination of sulforaphane and silibinin at each of the tested dosages had a synergistic effect compared to each component alone. For example, when the cells were treated with individual components, the glutathione levels remained the same with treatment of sulforaphane alone or decreased slightly with Silibinin treatment compared to the control. However, when cells were treated with the combination of sulforaphane and silibinin, at each of the tested dosages, the glutathione levels synergistically increased compared to the control. An increase in glutathione levels is a beneficial effect to detoxify cells. The results are depicted in FIG. 6.
  • Example 7
  • A subject presents with non-alcoholic fatty liver disease (NAFLD) and is suffering from symptoms including malaise, fatigue, and abdominal discomfort. She is administered a tablet containing glucoraphanin, myrosinase, ascorbic acid, and a milk thistle extract. The tablet is an enteric coated formulation which releases the contents in the small intestine. After one month of daily administration of the tablet, the subject experiences modulation of surrogate biomarkers including glutathione which correlates with improvement in symptoms.

Claims (14)

1. An orally administrable composition comprising:
a sulforaphane precursor;
an enzyme capable of converting the sulforaphane precursor to sulforaphane;
an enzyme potentiator; and
a milk thistle extract or powder.
2. The orally administrable composition of claim 1, wherein the sulforaphane precursor comprises glucoraphanin.
3. The orally administrable composition of claim 1, wherein the enzyme capable of converting the sulforaphane precursor to sulforaphane comprises myrosinase.
4. The orally administrable composition of claim 1, wherein the enzyme potentiator comprises ascorbic acid.
5. The orally administrable composition of claim 1, wherein the composition comprises an enteric-coated dosage form.
6. The orally administrable composition of claim 1, further comprising one or more additional components selected from the group consisting of: quercetin, an aminosugar, a glycosaminoglycan, avocado/soybean unsaponifiable, a vitamin, coffee fruit, magnesium, silymarin, proanthocyanidins, ursolic acid, curcumin, phytosterols, and phytostanols.
7. The orally administrable composition of claim 1, comprising glucoraphanin, myrosinase, ascorbic acid, and milk thistle extract.
8. The orally adminsitrable composition of claim 1, wherein the composition comprises broccoli extract or powder.
9. A method of treating, preventing, reducing the occurrence of, decreasing the symptoms associated with, and reducing secondary recurrences of rectal cancer or colon cancer, comprising administering to a subject in need thereof a sulforaphane precursor; an enzyme capable of converting the sulforaphane precursor to sulforaphane; an enzyme potentiator; and a milk thistle extract or powder.
10. The method of claim 9, wherein the sulforaphane precursor comprises glucoraphanin.
11. The method of claim 9, wherein the enzyme capable of converting the sulforaphane precursor to sulforaphane comprises myrosinase.
12. The method of claim 9, wherein the enzyme potentiator comprises ascorbic acid.
13. The method of claim 9, comprising administration of glucoraphanin, myrosinase, ascorbic acid, and milk thistle extract.
14. The method of claim 9, comprising administering an enteric-coated dosage form.
US14/586,704 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder Abandoned US20150118306A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/586,704 US20150118306A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US201261668328P 2012-07-05 2012-07-05
US201261668342P 2012-07-05 2012-07-05
US201261668396P 2012-07-05 2012-07-05
US201261668374P 2012-07-05 2012-07-05
US201261668386P 2012-07-05 2012-07-05
US201261668364P 2012-07-05 2012-07-05
US201361794417P 2013-03-15 2013-03-15
PCT/US2013/049261 WO2014008361A2 (en) 2012-07-05 2013-07-03 Compositions comprising a sulforaphane or a sulforaphane precursor and milk thistle extract or powder
US14/586,704 US20150118306A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/049261 Continuation WO2014008361A2 (en) 2012-07-05 2013-07-03 Compositions comprising a sulforaphane or a sulforaphane precursor and milk thistle extract or powder

Publications (1)

Publication Number Publication Date
US20150118306A1 true US20150118306A1 (en) 2015-04-30

Family

ID=49882601

Family Applications (14)

Application Number Title Priority Date Filing Date
US14/412,189 Abandoned US20150174093A1 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and magnesium
US14/412,176 Active US10688158B2 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder
US14/412,180 Abandoned US20150174213A1 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and ursolic acid
US14/412,191 Active 2035-02-15 US10960057B2 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US14/586,698 Abandoned US20150118305A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and ursolic acid
US14/586,704 Abandoned US20150118306A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder
US14/586,711 Abandoned US20150118304A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and magnesium
US14/586,765 Active US9421183B2 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US15/244,374 Active US10583178B2 (en) 2012-07-05 2016-08-23 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US16/701,644 Active US11654186B2 (en) 2012-07-05 2019-12-03 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US16/865,529 Active US11224639B2 (en) 2012-07-05 2020-05-04 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder
US17/007,067 Pending US20210008176A1 (en) 2012-07-05 2020-08-31 Compositions comprising sulforaphane or a sulforaphane precursor and magnesium
US17/178,664 Pending US20210169997A1 (en) 2012-07-05 2021-02-18 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US17/560,494 Pending US20220118062A1 (en) 2012-07-05 2021-12-23 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US14/412,189 Abandoned US20150174093A1 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and magnesium
US14/412,176 Active US10688158B2 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder
US14/412,180 Abandoned US20150174213A1 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and ursolic acid
US14/412,191 Active 2035-02-15 US10960057B2 (en) 2012-07-05 2013-07-03 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US14/586,698 Abandoned US20150118305A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and ursolic acid

Family Applications After (8)

Application Number Title Priority Date Filing Date
US14/586,711 Abandoned US20150118304A1 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and magnesium
US14/586,765 Active US9421183B2 (en) 2012-07-05 2014-12-30 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US15/244,374 Active US10583178B2 (en) 2012-07-05 2016-08-23 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US16/701,644 Active US11654186B2 (en) 2012-07-05 2019-12-03 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US16/865,529 Active US11224639B2 (en) 2012-07-05 2020-05-04 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder
US17/007,067 Pending US20210008176A1 (en) 2012-07-05 2020-08-31 Compositions comprising sulforaphane or a sulforaphane precursor and magnesium
US17/178,664 Pending US20210169997A1 (en) 2012-07-05 2021-02-18 Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
US17/560,494 Pending US20220118062A1 (en) 2012-07-05 2021-12-23 Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder

Country Status (11)

Country Link
US (14) US20150174093A1 (en)
EP (5) EP2849737B1 (en)
JP (10) JP6436443B2 (en)
CN (2) CN108355136B (en)
AU (8) AU2013286713B2 (en)
CA (5) CA2877329C (en)
ES (3) ES2700223T3 (en)
PL (2) PL3409280T3 (en)
RU (2) RU2666953C2 (en)
TW (6) TW201402128A (en)
WO (4) WO2014008361A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140275235A1 (en) * 2013-03-15 2014-09-18 Loic Pierre Deleyrolle Treatment of Proliferative Disorders
EP3123874A1 (en) * 2015-07-28 2017-02-01 Fundacíon Tecnalia Research & Innovation Formulations comprising glucosinolates and myrosinase
US11020372B2 (en) 2015-03-24 2021-06-01 University Of Florida Research Foundation, Incorporated Dietary and natural product management of negative side effects of cancer treatment
US11666549B2 (en) 2013-03-14 2023-06-06 University Of Florida Research Foundation, Incorporated Regulation of cancer using natural compounds and/or diet

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10869853B2 (en) * 2011-10-31 2020-12-22 The Johns Hopkins University Compositions and methods for treating autism spectrum disorders
AU2013286713B2 (en) 2012-07-05 2018-04-05 Nutramax Laboratories, Inc. Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder
EP2968990A4 (en) * 2013-03-15 2016-08-24 Nutramax Lab Inc Sulforaphane/sulforaphane precursor and phytosterol/phytostanol compositions
KR101515573B1 (en) * 2014-11-07 2015-04-28 주식회사 파미니티 Functional composition containing extract of new green for detoxification
WO2016102177A1 (en) 2014-12-22 2016-06-30 Unilever Plc Hair composition
CN107614062A (en) 2015-03-12 2018-01-19 加利福尼亚大学董事会 With the method for ROR gamma inhibitors treating cancers
BR112017027836A2 (en) * 2015-06-26 2018-09-04 Univ Florida Method for treating inflammation using natural compounds and / or diet
CA2990566A1 (en) 2015-06-26 2016-12-29 Plant Bioscience Limited Glucoraphnin for use in the treatment and/or prevention of diabetes mellitus
WO2017013239A1 (en) * 2015-07-22 2017-01-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Nrf2 activators for the treatment of mycobacterial infections
GB201605013D0 (en) * 2016-03-24 2016-05-11 Inst Of Food Res S-methylcysteine sulfoxide for prostate cancer treatment
US11357250B2 (en) 2016-08-15 2022-06-14 Summit Innovation Labs LLC Treatment and prevention of diabetes and obesity
US11344575B2 (en) * 2016-08-15 2022-05-31 Summit Innovation Labs, LLC Vascular calcification prevention and treatment
WO2018124258A1 (en) * 2016-12-28 2018-07-05 株式会社 Pal Therapeutic agent for chronic fatigue syndrome
JP7057070B2 (en) * 2017-06-21 2022-04-19 カゴメ株式会社 Powdered foods, methods for producing them, and methods for promoting myrosinase activity in powdered foods.
GB2607844B (en) * 2017-09-28 2023-04-05 Commw Scient Ind Res Org Isothiocyanate containing Brassicaceae products and method of preparation thereof
CA3092802A1 (en) * 2018-03-05 2019-09-12 Laila Nutraceuticals Synergistic herbal compositions for the treatment of obesity and overweight
CN116474079A (en) * 2019-05-08 2023-07-25 深圳福山生物科技有限公司 Composition containing glucoraphanin and application thereof
WO2022047212A1 (en) * 2020-08-27 2022-03-03 Arizona Board Of Regents On Behalf Of The University Of Arizona Compositions and methods for treating neurodegenerative disorders
WO2022140409A1 (en) 2020-12-23 2022-06-30 Church & Dwight Co., Inc. Compositions and methods to increase production of isothiocyanates
CN112574161A (en) * 2021-01-19 2021-03-30 江苏德和生物科技有限公司 Preparation method of EGC-rich non-ester tea polyphenol
JP7288474B2 (en) 2021-03-10 2023-06-07 プライムプラネットエナジー&ソリューションズ株式会社 Method for producing non-aqueous electrolyte secondary battery, and negative electrode active material
KR20240038929A (en) 2021-05-26 2024-03-26 뉴트라맥스 래보러토리즈, 인크. Composition comprising sulforaphane or sulforaphane precursor and moringa plant components
US20230364133A1 (en) * 2022-05-10 2023-11-16 Melaleuca, Inc. Dietary supplement compositions

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5725895A (en) * 1995-09-15 1998-03-10 Johns Hopkins School Of Medicine Method of preparing a food product from cruciferous seeds
US20030091518A1 (en) * 1999-12-20 2003-05-15 Gilles Pauly Cosmetic and/or pharmaceutical preparations
US20060003947A1 (en) * 2005-06-03 2006-01-05 Udell Ronald G Soft gel capsules containing polymethoxylated flavones and palm oil tocotrienols
US7402569B2 (en) * 2000-08-21 2008-07-22 Brassica Foundation For Chemoprotection Research, Inc. Treatment of Helicobacter with isothiocyanates

Family Cites Families (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3442639A1 (en) 1984-11-22 1986-05-22 Dr. Madaus & Co, 5000 Köln FLAVOLIGNANE DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
US5032401A (en) * 1989-06-15 1991-07-16 Alpha Beta Technology Glucan drug delivery system and adjuvant
US5223491A (en) 1989-11-09 1993-06-29 Donzis Byron A Method for revitalizing skin by applying topically water insoluble glucan
US5411986A (en) 1993-03-12 1995-05-02 The Johns Hopkins University Chemoprotective isothiocyanates
JPH07252148A (en) * 1994-10-08 1995-10-03 Masahiro Nagahama Therapeutic agent for diabetes
US5576015A (en) 1995-03-02 1996-11-19 Donzis; Byron A. Substantially purified beta (1,3) finely ground yeast cell wall glucan composition with dermatological and nutritional uses
US5686108A (en) * 1995-09-27 1997-11-11 Amway Corporation Brassica vegetable supplement and process for manufacture
JP3041232B2 (en) * 1995-11-16 2000-05-15 日本ケミカルリサーチ株式会社 Cancer metastasis inhibitor
JP2918834B2 (en) 1995-12-25 1999-07-12 美穂 田中 Production method of shiitake extract powder
JP2859843B2 (en) 1996-03-08 1999-02-24 株式会社雪国まいたけ Antitumor substance extracted from Maitake
US6242018B1 (en) 1997-04-11 2001-06-05 Johns Hopkins School Of Medicine Cancer Chemoprotective food products
US20010000472A1 (en) 1998-02-27 2001-04-26 Nutramax Laboratories, Inc. L-ergothioneine, milk thistle, and s-adenosylmethionine for the prevention, treatment and repair of liver damage
AU3308999A (en) 1998-02-27 1999-09-15 Nutramax Laboratories, Inc. L-ergothioneine, milk thistle, and s-adenosylmethionine for the prevention, treatment and repair of liver damage
US6521818B1 (en) 1998-07-01 2003-02-18 John Hopkins School Of Medicine Development of novel highly chemoprotectant crucifer germplasm
EP1100517A1 (en) * 1998-08-04 2001-05-23 John V. Kosbab Nutrient and therapeutic compositions for the treatment of cancer
JP4308350B2 (en) 1998-11-27 2009-08-05 小林製薬株式会社 LAK activity screening substance containing shiitake mycelium extract and LAK activity screening method using the same
AU2992901A (en) 2000-01-31 2001-08-07 University Of British Columbia, The Method for preparing and administering medicinal plant material
US6511675B2 (en) * 2000-06-12 2003-01-28 Access Business Group International, Llc Composition and method for correcting a dietary phytochemical deficiency
JP3504612B2 (en) 2000-12-28 2004-03-08 株式会社東洋新薬 Dietary supplement
US6582723B2 (en) 2001-05-03 2003-06-24 Wayne F. Gorsek Cancer immune composition for prevention and treatment of individuals
US8017160B2 (en) * 2003-08-15 2011-09-13 Russell Jaffe Enhancement of magnesium uptake in mammals
JP2005073508A (en) 2003-08-28 2005-03-24 Asahi Denka Kogyo Kk Beverage comprising edible or medicinal plant
ITMI20040696A1 (en) 2004-04-08 2004-07-08 Aboca S P A COMPOSITIONS FOR HERBAL TEA ENRICHED WITH VEGETABLE DRIED EXTRACTS
JP4126053B2 (en) * 2004-05-06 2008-07-30 堯 近藤 Method for producing health food containing Kazuno Reishi
WO2005107496A1 (en) 2004-05-06 2005-11-17 Maruasaen Corporation Method of producing fresh mushroom extract, extract and extract-containing material
RU2292899C2 (en) 2004-06-29 2007-02-10 Дмитрий Николаевич Мясников Method and remedy for preventing and reducing unfavorable manifestations of acute alcohol intoxication (versions) and method for producing the remedy
CA2603235A1 (en) * 2005-03-28 2006-10-05 Bioresponse, Llc Diindolylmethane-based compositions and methods of use thereof for promoting oral mucosal and bone health
EP1709969A1 (en) 2005-04-07 2006-10-11 Praktijkonderzoek Plant en Omgeving B.V. Health promoting dairy and food products containing mushroom glucan produced through fermentation of Grifola frondosa
KR100661032B1 (en) * 2005-04-19 2006-12-22 주식회사한국야쿠르트 Composition for Improving Liver Function, Reduction of Serum Ethanol Level, and Antioxidation
US20090247477A1 (en) * 2005-04-29 2009-10-01 John Hopkins School Of Medicine Methods of suppressing uv light-induced skin carcinogenesis
WO2006127903A1 (en) 2005-05-24 2006-11-30 Vdf Futureceuticals, Inc. Compositions and methods for reduction of ldl oxidation
DE102005033616A1 (en) * 2005-07-19 2007-01-25 Biopro Ag Biological Products Production of Brassica extracts with high anti-cancer sulforaphane content involves degradation of plant material in presence of myrosinase enzyme and glucosinolate
US20070021376A1 (en) * 2005-07-21 2007-01-25 Suracell, Inc. Supplement composition and method of use in enhancement of methylation process
US7597910B2 (en) 2005-08-20 2009-10-06 Slgm Medical Research Institute Compositions and methods for treating prostate disorders
RU2319494C2 (en) * 2005-11-11 2008-03-20 РАНХЕЛЬ Хосе Анхель ОЛАЛДЕ Synergistic composition of vegetable origin (variants), method for treatment of diseases with its using
US8323644B2 (en) 2006-01-17 2012-12-04 Sloan-Kettering Institute For Cancer Research Therapy-enhancing glucan
CN101553134A (en) * 2006-02-10 2009-10-07 曼纳泰克公司 All natural multivitamin and multimineral dietary supplement formulations for enhanced absorption and biological utilization
JP2007320947A (en) * 2006-05-30 2007-12-13 Shonan Institute For Medical & Preventive Science Agent for suppressing rise in blood sugar level
TW200806304A (en) 2006-06-02 2008-02-01 Heimat Co Ltd Maitake mushroom extract and composition containing the same for enhance production of sebum
WO2008002175A1 (en) * 2006-06-29 2008-01-03 Vulf Abramovich Laskin Weekly diet food package for oncological patients and for long-living persons
US7803605B2 (en) 2006-07-14 2010-09-28 National Institute Of Advanced Industrial Science And Technology Breeding method for yeast, yeast and a production method for glycoprotein or beta-glucan
WO2008115583A1 (en) * 2007-03-21 2008-09-25 John Mini Herbal treatments
US20080254055A1 (en) 2007-04-11 2008-10-16 John Erich Oblong Compositions for Regulation of Hair Growth
US20080311192A1 (en) * 2007-06-12 2008-12-18 Kraft Foods Holdings, Inc. Enteric-Coated Glucosinolates And Beta-Thioglucosidases
WO2009008006A2 (en) 2007-07-06 2009-01-15 Lupin Limited Pharmaceutical compositions for gastrointestinal drug delivery
CZ305673B6 (en) 2007-08-02 2016-02-03 Irel, Spol. S R. O. Mechanically treated milk thistle fruit and a method of its treatment
ES2325291B1 (en) 2007-10-04 2010-04-22 Madaus S A "USE OF A SILYBUM MARIANUM EXTRACT"
JP2011500680A (en) 2007-10-16 2011-01-06 ジョンズ・ホプキンス・ユニヴァーシティ How to protect skin from radiation injury
WO2009063885A1 (en) 2007-11-13 2009-05-22 Heimat Co., Ltd. Grifola frondosa extract and composition containing the same for promoting the production of hyaluronic acid (hyaluronan)
ITMI20080172A1 (en) * 2008-02-05 2009-08-06 Bios Line Spa ORAL FORMULATIONS FOR PROTECTION OF RESPIRATORY PATHWAYS WITH SPECIAL REFERENCE TO INFLAMMATORY AND NEOPLASTIC PHENOMENA
US20090252758A1 (en) * 2008-04-07 2009-10-08 Mazed Mohammad A Nutritional supplement for the prevention of cardiovascular disease, alzheimer's disease, diabetes, and regulation and reduction of blood sugar and insulin resistance
US7923044B2 (en) * 2008-07-15 2011-04-12 Paradise Herbs & Essentials, Inc. Composition for high-ORAC value dietary supplement
KR20100016876A (en) * 2008-08-05 2010-02-16 오춘근 A mixed grain for diet and glycosuria
EP2213280A1 (en) 2009-01-30 2010-08-04 DSM IP Assets B.V. Formulations comprising glucosinolate and myrosinase
CN101514174B (en) * 2009-02-24 2011-11-02 黑龙江八一农垦大学 Method for extracting multifunctional sulforaphane from broccoli sprouting vegetable
US8828953B2 (en) * 2009-04-20 2014-09-09 NaZura BioHealth, Inc. Chemosensory receptor ligand-based therapies
JP2010259424A (en) * 2009-05-11 2010-11-18 Hisako Arai Method for cooking supplementary dish easily taken in
US8377473B2 (en) 2009-07-01 2013-02-19 Magceutics, Inc. Slow release magnesium composition and uses thereof
WO2011060585A1 (en) * 2009-11-20 2011-05-26 天津天狮生物发展有限公司 Chewable tablets including essences of five-colour fruits and vegetables and preparative methods thereof
WO2011076154A1 (en) 2009-12-22 2011-06-30 Irel, Spol. S R.O. Feed supplement based on milk thistle, method of its production and its use
KR101079643B1 (en) 2010-01-12 2011-11-04 이현재 Composition of food for augmenting kidney function and preparation method thereof
WO2011099665A1 (en) 2010-02-12 2011-08-18 주식회사 케이씨아이 Antimicrobial composition containing extracts from natural ingredients, natural composite antiseptics, and method for manufacturing same
US20110206721A1 (en) * 2010-02-19 2011-08-25 Vijaya Nair Fermented soy nutritional supplements including mushroom components
US20120021079A1 (en) * 2010-02-23 2012-01-26 Brett Justin West Garcinia Mangostana L. and Iridoid Based Formulations
WO2011149835A1 (en) * 2010-05-24 2011-12-01 Max International, Llc Compositions and beverages comprising nutrients, vitamins, sugars, cysteine, and/or sugar-cysteine products
DE102010022587A1 (en) * 2010-05-28 2011-12-01 Wilfried Rühle Gastric juice-resistant composition for oral ingestion of plant-based components, useful for preventing colorectal cancer, comprises glucosinolate, and a construct of glucan-melanin-chitin complex coated with plant-based component
CN102450534A (en) * 2010-10-26 2012-05-16 宁波海逸生物科技有限公司 Formula of Xiling capsule with immunity enhancement function
ES2382299B1 (en) 2010-11-11 2013-05-07 Consejo Superior De Investigaciones Científicas (Csic) VEGETABLE POWDER FOR FOOD AND VEGETABLE PROTECTION AND PREPARATION METHODS.
NZ589578A (en) * 2010-11-29 2013-03-28 Comvita Ltd Cancer Chemoprotective Product including glucosinolate and myrosinase
CN102526455A (en) 2010-12-16 2012-07-04 天津中敖生物科技有限公司 Livestock Chinese medicinal composition with lung-clearing and phlegm-eliminating functions and preparation method thereof
US10925934B2 (en) * 2011-02-22 2021-02-23 Caudill Seed and Warehouse Co., Inc. Spray dried myrosinase and use to produce isothiocynates
WO2012122295A2 (en) * 2011-03-07 2012-09-13 Ned Biosystems, Inc. Treatment for pancreatic adenocarcinoma and other cancers of epithelial origin
US20130045273A1 (en) * 2011-08-19 2013-02-21 John Cuomo Methods for using nutritional supplements containing lipoic acids and sulfur containing compounds
CN102526445B (en) * 2011-11-07 2013-12-11 宁波海逸生物科技有限公司 Health-care medicament formula with functions of strengthening immunity, relieving physical fatigue and resisting tumor
AU2013286713B2 (en) * 2012-07-05 2018-04-05 Nutramax Laboratories, Inc. Compositions comprising sulforaphane or a sulforaphane precursor and a mushroom extract or powder

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5725895A (en) * 1995-09-15 1998-03-10 Johns Hopkins School Of Medicine Method of preparing a food product from cruciferous seeds
US5725895B1 (en) * 1995-09-15 2000-10-10 Hopkins J School Of Medicine Method of preparing food product from cruciferous seeds
US20030091518A1 (en) * 1999-12-20 2003-05-15 Gilles Pauly Cosmetic and/or pharmaceutical preparations
US7402569B2 (en) * 2000-08-21 2008-07-22 Brassica Foundation For Chemoprotection Research, Inc. Treatment of Helicobacter with isothiocyanates
US20060003947A1 (en) * 2005-06-03 2006-01-05 Udell Ronald G Soft gel capsules containing polymethoxylated flavones and palm oil tocotrienols

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Knoll et al. (Cancer Research, Published May 2005, pages 4448-4457) *
Shankar et al. (Clinical Cancer Research, Published November 1, 2008, Pages 6855-6866) *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11666549B2 (en) 2013-03-14 2023-06-06 University Of Florida Research Foundation, Incorporated Regulation of cancer using natural compounds and/or diet
US20140275235A1 (en) * 2013-03-15 2014-09-18 Loic Pierre Deleyrolle Treatment of Proliferative Disorders
US11020372B2 (en) 2015-03-24 2021-06-01 University Of Florida Research Foundation, Incorporated Dietary and natural product management of negative side effects of cancer treatment
EP3123874A1 (en) * 2015-07-28 2017-02-01 Fundacíon Tecnalia Research & Innovation Formulations comprising glucosinolates and myrosinase
WO2017016906A1 (en) * 2015-07-28 2017-02-02 Fundación Tecnalia Research & Innovation Formulation of glucosinolates and myrosinase

Also Published As

Publication number Publication date
TWI606830B (en) 2017-12-01
EP3409280A1 (en) 2018-12-05
TW201402129A (en) 2014-01-16
JP2019023245A (en) 2019-02-14
US20150196525A1 (en) 2015-07-16
EP2849737A4 (en) 2016-07-13
JP7232806B2 (en) 2023-03-03
CN104427981A (en) 2015-03-18
US10583178B2 (en) 2020-03-10
RU2666953C2 (en) 2018-09-13
TW201618791A (en) 2016-06-01
CA3112680A1 (en) 2014-01-09
EP3821895A1 (en) 2021-05-19
JP6777366B2 (en) 2020-10-28
RU2015103498A (en) 2016-08-27
WO2014008366A3 (en) 2014-03-13
US20150118304A1 (en) 2015-04-30
US10960057B2 (en) 2021-03-30
AU2020217440A1 (en) 2020-09-03
AU2018204604C1 (en) 2020-11-19
AU2018204604A1 (en) 2018-07-12
JP7458443B2 (en) 2024-03-29
US10688158B2 (en) 2020-06-23
US20150147352A1 (en) 2015-05-28
PL3409280T3 (en) 2021-10-18
JP7109132B2 (en) 2022-07-29
CA2877356C (en) 2021-05-11
JP2021038258A (en) 2021-03-11
TW201717964A (en) 2017-06-01
EP2849736B1 (en) 2018-09-05
WO2014008341A2 (en) 2014-01-09
WO2014008366A2 (en) 2014-01-09
JP2020128416A (en) 2020-08-27
JP2015523366A (en) 2015-08-13
RU2018130094A (en) 2018-10-04
ES2700223T3 (en) 2019-02-14
EP2849737B1 (en) 2019-09-11
US20200101143A1 (en) 2020-04-02
CA3112680C (en) 2023-07-25
WO2014008361A2 (en) 2014-01-09
AU2022203801A1 (en) 2022-06-23
CA2877338C (en) 2022-10-11
US11224639B2 (en) 2022-01-18
AU2013286713A1 (en) 2015-01-22
AU2013286721A1 (en) 2015-01-22
TWI580425B (en) 2017-05-01
JP7068383B2 (en) 2022-05-16
US20150110872A1 (en) 2015-04-23
RU2018130094A3 (en) 2022-04-19
JP2022137226A (en) 2022-09-21
JP2024023619A (en) 2024-02-21
PL2849736T3 (en) 2019-06-28
US20220118062A1 (en) 2022-04-21
CA2877338A1 (en) 2014-01-09
US20210169997A1 (en) 2021-06-10
EP2849736A4 (en) 2016-07-13
TWI637745B (en) 2018-10-11
AU2018204283A1 (en) 2018-07-05
US20210008176A1 (en) 2021-01-14
CA2877393C (en) 2020-01-07
WO2014008353A2 (en) 2014-01-09
TW201402130A (en) 2014-01-16
AU2020223773B2 (en) 2023-04-27
US20150174093A1 (en) 2015-06-25
TW201402131A (en) 2014-01-16
AU2018204604B2 (en) 2020-06-04
AU2023208131A1 (en) 2023-08-17
WO2014008361A3 (en) 2014-02-20
CA2877329C (en) 2020-01-07
RU2015103499A (en) 2016-08-27
CA2877356A1 (en) 2014-01-09
JP6548573B2 (en) 2019-07-24
JP6436443B2 (en) 2018-12-12
TW201402128A (en) 2014-01-16
WO2014008353A3 (en) 2014-03-06
US11654186B2 (en) 2023-05-23
AU2020217440B2 (en) 2022-09-08
CN104427981B (en) 2018-05-25
JP2015522039A (en) 2015-08-03
AU2013286721B2 (en) 2018-04-26
CA2877329A1 (en) 2014-01-09
EP2849737A2 (en) 2015-03-25
CA2877393A1 (en) 2014-01-09
WO2014008341A3 (en) 2014-02-27
US20150174213A1 (en) 2015-06-25
US20180200344A9 (en) 2018-07-19
EP3666277A1 (en) 2020-06-17
AU2020223773A1 (en) 2020-09-17
AU2018204283B2 (en) 2020-07-09
ES2755757T3 (en) 2020-04-23
EP2849736A2 (en) 2015-03-25
ES2877326T3 (en) 2021-11-16
US20200261553A1 (en) 2020-08-20
RU2680387C2 (en) 2019-02-20
JP2019014763A (en) 2019-01-31
JP2021001229A (en) 2021-01-07
US20150118305A1 (en) 2015-04-30
JP2022059053A (en) 2022-04-12
CN108355136A (en) 2018-08-03
AU2013286713B2 (en) 2018-04-05
US20160354448A1 (en) 2016-12-08
EP3409280B1 (en) 2021-04-14
CN108355136B (en) 2021-12-07
US9421183B2 (en) 2016-08-23

Similar Documents

Publication Publication Date Title
US11224639B2 (en) Compositions comprising sulforaphane or a sulforaphane precursor and a milk thistle extract or powder
RU2785683C2 (en) Compositions containing sulforaphane or sulforaphane precursor and extract or powder of milk thistle

Legal Events

Date Code Title Description
AS Assignment

Owner name: NUTRAMAX LABORATORIES, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CORNBLATT, BRIAN;CORNBLATT, GRACE;BZHELYANSKY, ANTON;AND OTHERS;SIGNING DATES FROM 20150205 TO 20150423;REEL/FRAME:035558/0558

STCB Information on status: application discontinuation

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