WO2013175479A1 - Compositions comprising isosilybin b for amelioration and prevention of drug- induced toxicity - Google Patents

Compositions comprising isosilybin b for amelioration and prevention of drug- induced toxicity Download PDF

Info

Publication number
WO2013175479A1
WO2013175479A1 PCT/IL2013/050446 IL2013050446W WO2013175479A1 WO 2013175479 A1 WO2013175479 A1 WO 2013175479A1 IL 2013050446 W IL2013050446 W IL 2013050446W WO 2013175479 A1 WO2013175479 A1 WO 2013175479A1
Authority
WO
WIPO (PCT)
Prior art keywords
acetaminophen
nac
apap
drug
administration
Prior art date
Application number
PCT/IL2013/050446
Other languages
French (fr)
Inventor
Josef MOGRABI
Yaron Ilan
Original Assignee
Hadasit Medical Research Services And Development Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hadasit Medical Research Services And Development Ltd. filed Critical Hadasit Medical Research Services And Development Ltd.
Priority to EP13793975.7A priority Critical patent/EP2854798A4/en
Publication of WO2013175479A1 publication Critical patent/WO2013175479A1/en
Priority to US14/551,724 priority patent/US9474763B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • 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
    • A61K31/10Sulfides; Sulfoxides; Sulfones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic 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
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • 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
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • A61K31/685Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols one of the hydroxy compounds having nitrogen atoms, e.g. phosphatidylserine, lecithin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/7056Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing five-membered rings with nitrogen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics

Definitions

  • the present invention relates to the treatment and prevention of drug-induced toxicity, such as liver and kidney toxicity.
  • the present invention relates to compositions and methods useful in treating, reducing and/or preventing liver failure, liver injury, liver damage and other toxicity effects associated with acetaminophen or caused by acetaminophen overdose.
  • Acetaminophen (chemically named N-(4-hydroxyphenyl)acetamide), also known as para-acetylaminophenol, paracetamol and N-acetyl-para-aminophenol (abbreviated as APAP), is a widely used analgesic and antipyretic drug. It is commonly used for the relief of headaches and other minor pains, and is a major ingredient in various cold and flu remedies.
  • Acetaminophen is the generic name of a drug found in many common brand-name over-the-counter (OTC) and prescription (Rx) products.
  • OTC over-the-counter
  • Rx prescription
  • acetaminophen is the sole active ingredient (e.g., TYLENOL®), while in others it is combined with additional active ingredients, such as opioid analgesics, nasal decongestants, cough suppressants and antihistamines. Examples include combinations with oxycodone (e.g. PERCOCET®) and hydrocodone (e.g. VICODIN®).
  • Acetaminophen is available as a tablet, capsule, liquid suspension, suppository, intravenous, and intramuscular form.
  • acetaminophen-induced liver toxicity is the most prevalent cause of acute liver failure in the Western world.
  • drug-host interactions such as drug viral interactions were suggested as potential causes for acetaminophen-induced liver toxicity even without exceeding the recommended dosages.
  • NAPQI N-acetyl-p-benzo-quinone imine
  • NK and NKT cells were suggested as important factors in this context.
  • liver injury from acetaminophen overdose remains a serious public health problem.
  • acetaminophen In addition to the liver, renal damage is also associated with acetaminophen consumption. In toxic amounts, acetaminophen may result in acute tubular necrosis. At therapeutic dosages, acetaminophen can be toxic in patients with risk factors such as starvation and chronic ingestion of alcohol or certain medications. Long term consumption of normal doses of acetaminophen has also been found to result in cumulative damage to the kidneys.
  • the latter may result in the consumption of more than one acetaminophen containing product without realizing that acetaminophen is included in all of these products, and accordingly an increased risk of acetaminophen overdose.
  • it can be difficult to recognize and diagnose the onset of liver injury.
  • Current treatments of acetaminophen overdose are mainly aimed at removing acetaminophen from the body and trying to replace diminished glutathione levels.
  • Oral administration of activated charcoal can also be used to decrease absorption of acetaminophen by binding any drug remaining in the gastrointestinal tract.
  • the treatment also includes administration of an antidote, N-acetylcysteine (NAC), which is a known precursor for glutathione, thereby helping the body to regenerate reduced glutathione and prevent or reduce damage to the liver.
  • NAC N-acetylcysteine
  • a liver transplant is sometimes required if damage to the liver becomes severe and irreversible.
  • liver transplantation is the only way to prevent deterioration and death.
  • US 4,868,114 discloses a method comprising stimulating the biosynthesis of glutathione in mammalian cells by contacting the cells with an effective amount of cysteine prodrugs.
  • US 6,555,141 discloses therapeutic compositions for the protection, treatment and repair of liver tissue comprising two or more compounds selected from the group consisting of S-adenosylmethionine, L-ergothioneine, and a compound selected from the group consisting of Milk thistle (Silybum marianum), silymarin and active components of silymarin, whether naturally, synthetically, or semi-synthetically derived, and to methods of preventing and treating liver disease and of repairing damaged liver tissue.
  • two or more compounds selected from the group consisting of S-adenosylmethionine, L-ergothioneine and a compound selected from the group consisting of Milk thistle (Silybum marianum), silymarin and active components of silymarin, whether naturally, synthetically, or semi-synthetically derived, and to methods of preventing and treating liver disease and of repairing damaged liver tissue.
  • US 6,566,401 discloses pharmaceutical compositions for the treatment or prevention of the toxic effects of therapeutic agents and methods of treating or preventing such toxicity using a toxicity reducing amount of N-acetylcysteine either alone or in combination with a therapeutically effective amount or, to achieve its therapeutic advantages, an amount larger than what is customarily given as a therapeutically effective amount, of a therapeutic agent.
  • US 7,238,373 discloses a nutritional supplement that is designed to provide nutritional benefits as well as to assist the body with detoxification.
  • US 8,148,356 discloses acetylcysteine compositions in solution, comprising acetylcysteine and which are substantially free of metal chelating agents, such as EDTA. Further, this invention relates to methods of making and using the acetylcysteine compositions. The present compositions and methods are designed to improve patient tolerance and compliance, while at the same time maintaining the stability of the pharmaceutical formulation. The compositions and methods of this invention are useful in the treatment of acetaminophen overdose, acute liver failure, various cancers, methacrylonitrile poisoning, reperfusion injury during cardio bypass surgery, and radiocontrast-induced nephropathy, and can also be used as a mucolytic agent.
  • US 2011/0124718 discloses the reduction of acetaminophen toxicity by dietary milk thistle extract. Milk thistle and acetaminophen are combined to provide single dosages containing both ingredients, whether dispensed in a liquid suspension, chewable tablets, coated caplets, gelcaps, geltabs, and suppositories.
  • US 2012/0022161 discloses a pharmaceutical composition and method for providing a reduction in side effects for human patients in need of therapy comprising the administration of a pharmaceutical composition comprising acetylcysteine.
  • compositions comprising a combination of amino acids selected from: a) L-Cysteine with L-Methionine; b) L-Cysteine with L-Serine; c) L- Cysteine with L-Methionine and L-Serine; d) L-Methionine with L-Serine; or precursors or transporters of said amino acids, for the prevention of paracetamol-induced liver damage.
  • WO 2011/044230 discloses methods and compositions comprising N- acetylcysteine amide (NAC amide) and derivatives thereof for use in treatments and prophylactic therapies for human and non-human mammalian diseases, disorders, conditions and pathologies associated with bomb blast or other high energy noise or impulse blasts.
  • NAC amide N- acetylcysteine amide
  • Pharmaceutically or physiologically acceptable compositions of NAC amide or derivatives thereof are also provided, which may be administered alone, or in combination with other suitable agents.
  • the present invention provides pharmaceutical compositions comprising combinations of protective agents which are effective in reducing and/or preventing drug- induced toxicity, including but not limited to, acetaminophen-induced toxicity.
  • the protective agents utilized herein are selected from isosilybin B, methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno-cysteine (Se-Cys), ribose-cysteine (RibCys), N-acetylcysteine (NAC), N-acetylcysteine-amide (AD4), methionine (Met) and S-adenosylmethionine (SAM).
  • MSM methylsulfonylmethane
  • Cys phosphatidylcholine 5 cysteine
  • Se-Cys seleno-cysteine
  • RibCys ribose-cysteine
  • compositions containing the protective agents may be formulated with or without a drug or substance with potential toxicity, and accordingly may be used as safe formulations of the drug, or as an antidote in case of intoxication.
  • compositions are formulated with or without acetaminophen.
  • Acetaminophen-containing compositions may be used as safe formulations with reduced risk of causing liver damage and/or renal damage.
  • Compositions without acetaminophen can be used as an antidote for the treatment of acetaminophen-related toxicity, such as liver failure or liver disorder.
  • the present invention further provides methods for treating drug-induced intoxication, such as acetaminophen-induced intoxication, utilizing the combinations of protective agents disclosed herein.
  • the combinations of protective agents of the present invention were found to be remarkably effective in reducing liver damage and liver toxicity associated with acetaminophen administration.
  • mice exposed to sub-lethal amounts of acetaminophen administered with different combinations of the above compounds showed significantly reduced serum levels of liver enzymes and better body weight recovery following fast compared to mice exposed to same amounts of acetaminophen alone.
  • histological examination revealed little or no liver damage in mice that received the different combinations of protective compounds, in contrast to mice who received acetaminophen alone.
  • the ability of the compounds to protect the liver was demonstrated when the compounds and acetaminophen were administered concurrently, as well as when the compounds were administered several hours following acetaminophen administration.
  • the combinations of the present invention act at least additively and preferably synergistically and are superior to the currently used antidote (gold standard treatment) of acetaminophen poisoning, namely, NAC alone.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising isolated isosilybin B, and optionally further comprising at least one additional protective agent selected from the group consisting of methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno-cysteine (Se- Cys), ribose-cysteine (RibCys), N-acetylcysteine (NAC), N-acetylcysteine-amide (AD4), methionine (Met) and S-adenosylmethionine (SAM), wherein said pharmaceutical composition is effective in treating or preventing drug-induced toxicity.
  • MSM methylsulfonylmethane
  • Cys phosphatidylcholine 5 cysteine
  • Se- Cys seleno-cysteine
  • RibCys ribose-cysteine
  • NAC N-acetyl
  • isosilybin B is one of the components of an extract produced from milk thistle (Silybum marianum). This compound is utilized herein in an isolated form, meaning that it is substantially separated from other components of the extract.
  • the composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable diluent, excipient or carrier.
  • the phosphatidylcholine component comprises a majority of unsaturated fatty acids. In some embodiments, the phosphatidylcholine component comprises a majority of polyunsaturated fatty acids. In some embodiments, the polyunsaturated phosphatidylcholine comprises unsaturated fatty acids selected from the group consisting of linoleic acid, linolenic acid and oleic acid. In some embodiments, linoleic acid constitutes about 70% or more of the fatty acid content of the phosphatidylcholine.
  • the phosphatidylcholine component comprises 1, 2-dilinoleoylphosphatidylcholine (DLPC), with linoleic acid bound both in cl- and c2-positions of the molecule. In some embodiments, about 50% or more of the phosphatidylcholine molecules present within the composition are DLPC.
  • DLPC 2-dilinoleoylphosphatidylcholine
  • the pharmaceutical composition is used for the treatment of acetaminophen intoxication.
  • the protective agents described herein are formulated together with a drug that can potentially cause toxic effects, in a single pharmaceutical composition.
  • the pharmaceutical composition of the present invention further comprises a drug with potential toxicity.
  • the drug is acetaminophen.
  • the pharmaceutical composition is characterized by a reduced risk of acetaminophen-induced toxicity, such as acetaminophen-induced liver damage and/or nephrotoxicity, compared to formulations of acetaminophen that do not contain the protective agents as described herein.
  • Such compositions are contemplated to substantially retain the efficacy of the drug, e.g. acetaminophen, while significantly reducing or preventing drug-induced liver toxicity, liver damage and kidney damage.
  • the acetaminophen-containing pharmaceutical composition of the present invention further comprises at least one additional drug selected from the group consisting of an opioid analgesic, a decongestant, a cough suppressant, an antihistamine and an expectorant.
  • the opioid analgesic is selected from the group consisting of codeine, oxycodone, hydrocodone and propoxyphene.
  • the decongestant is selected from the group consisting of phenylephrine and pseudoephedrine.
  • the expectorant is guaiphenesin.
  • the cough suppressant is selected from the group consisting of dextromethorphan and codeine.
  • the antihistamine is selected from the group consisting of chlorpheniramine, phenyltoloxamine and doxylamine.
  • the pharmaceutical composition is formulated for systemic administration.
  • the pharmaceutical composition is formulated for a route of administration selected from the group consisting of oral, rectal, intravenous, intramuscular, intradermal, subcutaneous and intranasal.
  • the present invention provides a method for treating drug intoxication in a subject in need thereof, the method comprising administering to the subject isolated isosilybin B, and optionally at least one additional protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys) Se-Cys, RibCys, NAC, AD4, Met and SAM.
  • the method comprises administering to the subject isolated isosilybin B and further administering (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
  • the protective agents are within a single composition. In other embodiments of the present invention, the protective agents are within separate compositions. In some embodiments, the two or more protective agents are administered concurrently. In other embodiments, the two or more protective agents are administered sequentially. In some embodiments, the two or more protective agents are administered via the same route of administration. In other embodiments, the two or more protective agents are administered via different routes of administration.
  • administration is performed via a route of administration selected from the group consisting of oral, rectal, intravenous, intramuscular, intradermal, subcutaneous and intranasal. In some embodiments, the route of administration is selected from the group consisting of oral and intravenous.
  • the method of the present invention is applied to a subject who has consumed, or suspected of having consumed, large amounts of acetaminophen.
  • acetaminophen intoxication is acute intoxication. In other embodiments, acetaminophen intoxication is chronic intoxication.
  • the subject is human. In other embodiments, the subject is a non-human mammal.
  • the present invention provides a composition comprising isolated isosilybin B, and optionally at least one additional protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, for use in the treatment of drug intoxication.
  • additional protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, for use in the treatment of drug intoxication.
  • the drug is acetaminophen.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable diluent, excipient or carrier.
  • the pharmaceutical composition is formulated for administration via a route of administration selected from the group consisting of oral, rectal, intravenous and intramuscular.
  • Figure 1 Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus IB, compared to mice administered with acetaminophen only.
  • Figure 2 Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus PC, compared to mice administered with acetaminophen only.
  • Figure 3 Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus PC, compared to mice administered with acetaminophen only.
  • Figure 4 Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus MSM, compared to mice administered with acetaminophen only.
  • FIG. 6 Representative liver biopsy results of mice treated with acetaminophen, acetaminophen plus NAC, acetaminophen plus Cys, PC, MSM and IB, or acetaminophen plus Met, PC, MSM and IB.
  • Figure 7 Representative liver biopsy results of mice treated with acetaminophen, acetaminophen plus NAC, PC, MSM and IB, acetaminophen plus Cys, PC, MSM and IB, or acetaminophen plus Met, PC, MSM and IB.
  • FIG. 8 Representative liver biopsy results of mice treated with acetaminophen, acetaminophen plus NAC, acetaminophen plus Cys, or acetaminophen plus Met.
  • the present invention relates to treatment and prevention of drug-induced toxicity, particularly acetaminophen-induced toxicity, using protective active ingredients selected from the group consisting of isosilybin B, methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno-cysteine (Se-Cys), ribose-cysteine (RibCys), N- acetylcysteine (NAC), N-acetylcysteine-amide (AD4), methionine (Met) and S- adenosylmethionine (SAM).
  • a plurality of protective active compounds is used.
  • a plurality indicates at least two.
  • a "protective agent” or “protective compound” refers to a compound effective in treating or preventing drug-induced toxicity.
  • the term refers to a compound that serves to decrease hepatotoxicity and/or nephrotoxicity caused by the consumption of a drug.
  • drug is used herein to refer to a compound with a therapeutic activity, most often a manmade small organic molecule, that can potentially cause toxic effects, such as damage to the liver and possible other organs, for example, acetaminophen.
  • a drug is a molecule known to exhibit potential hepatotoxicity.
  • the phrase "pharmaceutical composition effective in treating or preventing drug-induced toxicity” indicates that the protective agents are present in the pharmaceutical composition in an amount that is effective to perform their utility.
  • the phrase indicates that the protective agents are present in the composition in an amount that is effective to significantly reduce or even completely inhibit toxic effects of the drug.
  • the phrase indicates that the amount of protective agents is effective in alleviating or even eliminating liver injury and/or other damage caused by the consumption of a drug.
  • drug-induced toxicity refers to damages to body tissues and organs resulting from the consumption of a drug, typically consumption of large amounts of the drug.
  • drug intoxication refers to a clinical condition resulting from the consumption of toxic amounts of a drug (e.g. acetaminophen) that damage the liver and/or other organs such as the kidneys.
  • a drug e.g. acetaminophen
  • the extent of liver damage, as well as improvement and reduction in liver damage as a result of utilizing the compositions and methods of the present invention, may be evaluated, for example, by measuring serum levels of liver enzymes. An increased level of liver enzymes in the serum indicates liver damage. Nephrotoxicity can be monitored, for example, through simple blood and urine tests. A decreased glomerular filtration rate (GFR) indicates renal damage and poor renal function.
  • GFR glomerular filtration rate
  • the term "preventing”, when referring to drug-induced toxicity, encompasses significantly reducing the risk of toxicity or even completely inhibiting toxic effects of the drug.
  • the term "about”, when referring to a measurable value such as an amount, is meant to encompass variations of +/-10%, more preferably +1-5%, even more preferably +/-1 %, and still more preferably +/-0.1 % from the specified value, as such variations are appropriate to achieve the intended purpose.
  • compositions of the present invention comprise protective agents selected from isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM.
  • protective agents selected from isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM.
  • Pharmaceutically acceptable salts of the protective agents described herein are also within the scope of the present invention.
  • the compositions may be formulated with or without a drug with potential toxicity, such as acetaminophen.
  • a pharmaceutical composition effective in treating or preventing drug-induced, such as acetaminophen-induced, liver toxicity, the composition comprising isolated isosilybin B , and optionally further comprising at least one more protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM.
  • a pharmaceutical composition comprising the combinations of protective agents disclosed herein, for use in the treatment of drug intoxication.
  • the drug is acetaminophen.
  • the pharmaceutical composition comprises isosilybin B.
  • isosilybin B is combined with at least one more protective agent selected from the group consisting of NAC, phosphatidylcholine, MSM, RibCys, AD4, Cys, Se-Cys, Met and SAM.
  • the pharmaceutical composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
  • the pharmaceutical composition comprises isolated isosilybin B, NAC and one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
  • the pharmaceutical composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
  • the pharmaceutical composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys and Met; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
  • a pharmaceutical composition effective in treating or preventing drug-induced toxicity comprises a plurality of protective agents selected from the group consisting of isolated isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM.
  • a plurality of protective agents selected from the group consisting of isolated isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM.
  • the composition comprises NAC and at least one more protective agent selected from the group consisting of phosphatidylcholine, MSM, isosilybin B.
  • the combination of NAC and at least one more protective agent selected from the above compounds permits lower amounts of NAC in order to achieve a therapeutic effect compared to the amount required for NAC alone.
  • the composition comprises (i) one or more protective agents selected from the group consisting of AD4, Cys, Se-Cys, RibCys, Met and SAM; and (ii) one or more protective agents selected from the group consisting of phosphatidylcholine, MSM, isosilybin B.
  • the composition comprises a combination of two or more protective agents selected from the group consisting of NAC, phosphatidylcholine, MSM, isosilybin B, RibCys and SAM.
  • the pharmaceutical composition comprises NAC and phosphatidylcholine as the protective agents.
  • the pharmaceutical composition comprises NAC in combination with MSM or SAM as the protective agents.
  • the pharmaceutical composition comprises NAC and isosilybin B as the protective agents.
  • the combination of NAC and at least one more protective agent selected from the above compounds permits lower amounts of NAC in order to achieve a therapeutic effect compared to the amount required for NAC alone.
  • the pharmaceutical composition does not include NAC.
  • the compositions contain the protective agents described herein, and do not contain a drug that can cause intoxication.
  • Such compositions are effective in treating drug-induced toxicity, and can be administered in case that drug-induced toxicity has occurred, in order to treat said toxicity.
  • such compositions can be administered following an overdose of a drug, which results in signs of intoxication, in order to treat the toxicity effects.
  • the pharmaceutical compositions of the present invention can be used as an antidote for the treatment of acetaminophen intoxication.
  • compositions of the present invention contain the protective agents described herein, and further contain a drug that can cause intoxication. Such compositions are characterized by a reduced risk of drug intoxication compared to compositions that contain the drug without the protective agents.
  • the compositions contain acetaminophen and possibly additional one or more drugs.
  • safe formulations of acetaminophen are provided, with significantly reduced risk of causing liver and renal damage, even when taken at large amounts.
  • Such compositions have several advantages, including a significantly reduced risk for overdose and significantly reduced need for liver transplantation following an overdose.
  • such compositions may be beneficial to populations at risk.
  • a pharmaceutical composition which comprises acetaminophen, isosilybin B and at least one more protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, said pharmaceutical composition is characterized by a reduced risk of drug intoxication compared to a pharmaceutical composition comprising acetaminophen without isosilybin B and the at least one more protective agent.
  • a pharmaceutical composition which comprises acetaminophen and further comprises a plurality of protective agents selected from the group consisting of isolated isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, said pharmaceutical composition is characterized by a reduced risk of drug intoxication compared to a pharmaceutical composition comprising acetaminophen without the plurality of protective agents.
  • NAC may be identified by CAS Registry Number 616-91-1
  • MSM may be identified by CAS Registry Number 67-71-0
  • isosilybin B may be identified by CAS Registry Number 142796-22-3
  • SAM may be identified by CAS Registry Number 29908-03-0
  • RibCys may be identified by CAS Registry Number 17087-36-4
  • AD4 may be identified by CAS Registry Number 38520-57-9
  • Cys may be identified by CAS Registry Number 52-90-4
  • Se-Cys may be identified by CAS Registry Number 10236-58-5
  • Met may be identified by CAS Registry Number 63-68-3.
  • Phosphatidylcholine is a phospholipid composed of a choline headgroup and glycerophosphoric acid with a variety of fatty acids.
  • the fatty acids may include saturated and unsaturated fatty acids.
  • the phosphatidylcholine component comprises mostly unsaturated fatty acids.
  • the fatty acid content of the phosphatidylcholine comprises at least one of linoleic acid, linolenic acid and oleic acid.
  • linoleic acid constitutes about 70% or more of the fatty acid content of the phosphatidylcholine.
  • the phosphatidylcholine component comprises 1, 2-dilinoleoylphosphatidylcholine (DLPC), with two linoleic acid molecules. In some embodiments, about 50% or more of the phosphatidylcholine molecules present within the composition are DLPC.
  • DLPC 2-dilinoleoylphosphatidylcholine
  • Isosilybin B (also known as isosilibinin B) is a flavonolignan that constitutes one of the components of the milk thistle (Silybum marianum) extract.
  • Traditional milk thistle extract is made from the plant seeds and consists of about 65-80% silymarin (a flavonolignan complex) and 20-35% fatty acids.
  • Silymarin is a complex mixture of polyphenolic molecules, including seven closely related flavonolignans, namely silybin A, silybin B, isosilybin A, isosilybin B, silychristin, isosilychristin and silydianin, and one flavonoid (taxifolin).
  • isosilybin B is highly more potent and effective in reducing acetaminophen-induced liver toxicity, compared to other compounds found in silymarin, or in any other milk thistle extract of any part of the plant.
  • the closely related silybin B and isosilybin A did not show a beneficial effect when administered to mice together with, or following, large dosage of acetaminophen.
  • Isosilybin B may be purified from an extract of the milk thistle. Isosilybin B is utilized herein in an isolated form, that is separated from the other ingredients of the extract.
  • isolated indicates at least about 80%, separated from other ingredients of the extract, preferably at least about 90%, more preferably at least about 95% separated from other ingredients of the extract. Synthetic Isosilybin B may also be used.
  • the NAC is substantially free of its oxidized form, di-n- acetylcysteine. It is preferred that the therapeutic agent, serially or co-administered, be in any form in which it is typically available and the composition should be prepared in a manner that substantially prevents oxidation of the NAC during preparation or storage.
  • the preparation and storage of the formulation is performed in such a way that the reduced form of NAC is the primary form administered to the patient. Maintaining NAC containing formulations in solid form is preferable for this purpose.
  • NAC containing formulations are preferably stored in a dark bottle that is vacuum sealed. Storage in cool dark environments is also preferred.
  • the determination of reduced and oxidized species present in a sample may be determined by various methods known in the art, for example with capillary electrophoresis, HPLC, etc. as described by Chassaing et al. (1999) J Chromatogr B Biomed Sci Appl 735(2):219-27.
  • the protective agents described herein are present in the composition in an amount which is effective for inducing the therapeutic effect.
  • Exemplary dosage ranges for the different active protective agents include doses ranging between about 0.05 - 10 mg/kg, for example between about 0.05-5 mg/kg, between about 0.05-1 mg/kg, between about 0.1 - 2.5 mg/kg, between about 0.5-2 mg/kg.
  • two or more protective agents when they are present in the composition, they can be formulated such that they are released concurrently. In other embodiments, they are formulated such that they are released sequentially. For example, in case there are two protective agents within a pharmaceutical composition, one can be formulated for immediate release, and the other for sustained release. Such formulations are known in the art.
  • the protective agents are formulated within liposomes.
  • the liposomes may be used as a delivery method.
  • the liposomes may be used for liver targeting.
  • One or more of the protective agents may be used to create the liposomes.
  • the pharmaceutical composition is a combination medication that further comprises acetaminophen.
  • the composition may also include additional one or more drugs, such opioid analgesics, decongestants, cough suppressants, antihistamines, expectorants or combinations thereof.
  • Information about combination medications containing acetaminophen and at least one more active ingredient can be found, for example, at the MedLinePlus website (www.nlm.nih.gov/medlineplus/).
  • Non-limiting examples of opioid analgesics include codeine, oxycodone, hydrocodone and propoxyphene.
  • Non-limiting examples of decongestants include phenylephrine, pseudoephedrine and guaiphenesin.
  • Non-limiting examples of cough suppressants include dextromethorphan and codeine.
  • Non-limiting examples of antihistamines include chlorphenir amine, phenyltoloxamine and doxylamine.
  • a non- limiting example of an expectorant is guaiphenesin.
  • the combination of the protective compounds described herein with acetaminophen does not interfere with the normal pharmaceutical activity of acetaminophen, and it maintains its pain relieving and fever reducing properties.
  • Acetaminophen is commercially available and may also be synthesized by methods well known in the art. An overview of synthetic processes of acetaminophen can be found, for example, in Anthony S. Travis (2007). "Manufacture and uses of the anilines: A vast array of processes and products". In Zvi Rappoport. The chemistry of Anilines Part 1. Wiley, p. 764; and Elmar Friderichs, Thomas Christoph, Helmut Buschmann (2005), "Analgesics and Antipyretics", Ullmann's Encyclopedia of Industrial Chemistry, Weinheim: Wiley- VCH
  • compositions of the present invention that comprise acetaminophen may find use as anti-pyretic and analgesic agents, and are suitable for medical indications treatable with acetaminophen alone.
  • An improvement in product safety is provided by the inclusion of the protective compounds as disclosed herein, which substantially prevents the possibility of accidental or inadvertent overdose.
  • compositions of the present invention which will be effective in the treatment of a particular condition will depend on the nature of the condition, and can be determined by standard clinical techniques. See, for example, Goodman and Oilman; The Physician's Desk Reference, Medical Economics Company, Inc., Oradell, N.J., 1995; and to Drug Facts and Comparisons, Facts and Comparisons, Inc., St. Louis, Mo., 1993.
  • the addition of the compounds as disclosed herein does not affect the therapeutic efficacy of acetaminophen, it is generally not necessary to adjust the dosage from what would ordinarily be administered for acetaminophen alone, and in fact the dose may be raised due to the increased safety of the present formulations.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease, and should be decided according to the judgment of the practitioner and each patient's circumstances.
  • the acetaminophen present in orally administrable solid unit doses will usually be at least about 80 mg (for pediatric doses), 325 mg, 500 mg and 650 mg.
  • Oral liquid dosage forms usually comprise at least about 100 mg/ml, 120 mg/2.5 ml, 120mg/5 ml, 160 mg/5 ml, 165 mg/5 ml, 325 mg/5 ml acetaminophen.
  • Suppositories are formulated in the manner well known in the art and usually comprise at least about 120 mg, 125 mg, 325 mg, 500 mg and 650 mg acetaminophen per dosage unit Typically, the composition further comprises a pharmaceutically acceptable diluent, excipient or carrier.
  • the term "pharmaceutically acceptable diluent, excipient, or carrier” refers to a diluent, excipient, or carrier that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered active agent.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols. Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences", Mack Publishing Co., Easton, PA, (Remington: The Science and Practice of Pharmacy, Gennaro, A., Lippincott, Williams & Wilkins, Philadelphia, Pa., 20th ed, 2000).
  • a “carrier” refers to any substance suitable as a vehicle for delivering of the agents or molecule of the present invention to a suitable in vivo or in vitro site.
  • carriers can act as a pharmaceutically acceptable excipient of a therapeutic composition of the present invention.
  • Carriers of the present invention include: (1) excipients or formularies that transport, but do not specifically target a molecule to a cell (referred to herein as non-targeting carriers); and (2) excipients or formularies that deliver a molecule to a specific site in a subject or a specific cell (i.e., targeting carriers).
  • non-targeting carriers examples include, but are not limited to water, phosphate buffered saline, Ringer's solution, dextrose solution, serum-containing solutions, Hank's solution, other aqueous physiologically balanced solutions, oils, esters and glycols.
  • Aqueous carriers can contain suitable auxiliary substances required to approximate the physiological conditions of the recipient, for example, by enhancing chemical stability and isotonicity.
  • compositions of the present invention can be sterilized by conventional methods.
  • Any suitable route of administration may be used for the composition of the present invention, including but not limited to local and systemic routes.
  • Systemic administration includes all enteral and all parenteral routes.
  • suitable administration routes include oral, rectal, transmucosal such as transnasal and buccal, intravenous, intramuscular, transdermal, subcutaneous, intradermal, intravitreal, intravesicular and inhalation routes.
  • the composition of the present invention is formulated for topical administration. In other embodiments, the composition is formulated for systemic administration.
  • compositions of the present invention may be formulated in conventional manners. The proper formulation is dependent upon the route of administration chosen.
  • compositions of the present invention are formulated for oral administration.
  • formulations for oral administration include tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Suitable carriers for oral administration are well known in the art.
  • Compositions for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries as desired, to obtain tablets or dragee cores.
  • Non-limiting examples of suitable excipients include fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol, cellulose preparations such as, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, and sodium carbomethylcellulose, and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • disintegrating agents such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate, may be added.
  • the active ingredients of the composition may be formulated in aqueous solutions, for example in physiologically compatible buffers including but not limited to Hank's solution, Ringer's solution, or physiological salt buffer.
  • Formulations for injection may be presented in unit dosage forms, for example, in ampoules, or in multi-dose containers with, optionally, an added preservative.
  • the compositions may be in the form of suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.
  • Non-limiting examples of suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters such as ethyl oleate, triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the active ingredients, to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, for example, a sterile, pyrogen-free, water-based solution, before use.
  • a suitable vehicle for example, a sterile, pyrogen-free, water-based solution, before use.
  • it is preferable to adjust the pH of the administered solution to that of the blood, namely, about pH 7.4.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the active ingredients are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluorome thane, dichloro-tetrafluoroethane, or carbon dioxide.
  • a suitable propellant for example, dichlorodifluoromethane, trichlorofluorome thane, dichloro-tetrafluoroethane, or carbon dioxide.
  • the dosage may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, for example, gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base, such as lactose or starch.
  • compositions of the present invention are formulated for rectal administration, for example, as suppositories or retention enemas, using, for example, conventional suppository bases such as cocoa butter or other glycerides.
  • the exact formulation, route of administration, and dosage can be chosen by the individual physician in view of the patient's condition.
  • the composition further comprises at least one additive useful in the pharmaceutical fields, including, but not limited to fats, emulsifiers and co- emulsifiers, hydrophilic or lipophilic gelling agents, colorants, fragrances, emollients, humectants, preservatives, vitamins, chelators, solvents, fillers, thickeners, hydrophilic and lipophilic filters, dyestuffs, neutralizers, penetration-enhancing agents and polymers.
  • at least one additive useful in the pharmaceutical fields including, but not limited to fats, emulsifiers and co- emulsifiers, hydrophilic or lipophilic gelling agents, colorants, fragrances, emollients, humectants, preservatives, vitamins, chelators, solvents, fillers, thickeners, hydrophilic and lipophilic filters, dyestuffs, neutralizers, penetration-enhancing agents and polymers.
  • Non-limiting examples of suitable fats include mineral oils, oils of animal origin (lanolin), synthetic oils (isopropyl myristate, octyldodecyl, isostearyl isostearate, decyl oleate or isopropyl palmitate), silicone oils (cyclomethicone or dimethicone) and fluorinated oils.
  • mineral oils oils of animal origin (lanolin)
  • synthetic oils isopropyl myristate, octyldodecyl, isostearyl isostearate, decyl oleate or isopropyl palmitate
  • silicone oils cyclomethicone or dimethicone
  • Fatty alcohol, fatty acids, waxes and gums, notably silicone gums and elastomers can also be used as fats.
  • Non-limiting examples of suitable emulsifiers and co-emulsifiers include polyglycerol fatty acid esters, sucrose fatty acid esters, sorbitane fatty acid esters, oxyethylene sorbitan fatty acid esters, PEG fatty alcohol ethers, glycerol fatty acid esters, alkyl sulphates, alkyl ether sulphates, alkyl phosphates, alkyl polyglucosides and dimethicone copolyols.
  • Non-limiting examples of suitable hydrophilic gelling include carboxyvinyl polymers (carbomer), acrylic copolymers such as acrylate/alkylacrylate copolymers, polyacrylamids, polysaccharides such as xanthan gum, guar gum, natural gums such as cellulose gum and derivatives, clays and 2-acrylamido-2-methylpropane acid copolymers.
  • Non-limiting examples of suitable lipophilic gelling agents include modified clays such as bentones, fatty acid metal salts, hydrophobic silica and ethylcellulose.
  • Non-limiting examples of suitable fillers include talc, kaolin, mica, serecite, magnesium carbonate, aluminum silicate and organic powders such as nylon.
  • Non-limiting examples of suitable dyestuffs include lipophilic dyes, hydrophilic dyes, pigments and mother-of-pearl commonly used in cosmetic or dermatological compositions, and their mixtures.
  • Non-limiting examples of suitable neutralizers include soda, triethanolamine, aminomethyl propanol and potassium hydroxide.
  • Non-limiting examples of suitable penetration enhancing agents include alcohols and glycols (ethanol and propylene glycol), ethoxydiglycol, alcohols and fatty acids (oleic acid), fatty acid esters and dimethyl isosorbide.
  • Non-limiting examples of preservatives compatible with cosmetic and pharmaceutical compositions include benzoic acid, its salts and esters, sorbic acid and its salts, parabens and their salts, triclosan, imidazolidinyl urea, phenoxyethanol, DMDM hydantoin, diazolidinyl urea and chlorphenesin.
  • the filters are UVA and UVB filters.
  • suitable UVA and UVB filters include organic filters such as benzophenone-3, butyl methoxydibenzoyl methane, octocrylene, octyl methoxycinnamate, 4-methylbenzylidene camphor, octyl salicylate, terephthalylidene dicamphor sulfonic acid and drometrizole trisiloxane, and non-organic filters such as titanium oxide and zinc oxide.
  • Non-limiting examples of suitable solvents include water, ethanol, glycerin, propylene glycol, butylene glycol and sorbitol.
  • suitable solvents include water, ethanol, glycerin, propylene glycol, butylene glycol and sorbitol.
  • the quantities of these various additives are those conventionally used in pharmaceutical preparations as is known to a person skilled in the art.
  • a method for treating acetaminophen intoxication in a subject in need thereof comprising administering the combinations of protective agents disclosed herein.
  • isolated isosilybin B alone or in combination with one or more one more protective agents selected from the group consisting of MSM, phosphatidylcholine ⁇ Cys) Se-Cys, RibCys, NAC, AD4, Met and SAM are administered.
  • two or more protective agents are administered, selected from the group consisting of NAC, AD4, Cys, Se-Cys, Met, RibCys phosphatidylcholine, MSM, and isosilybin B.
  • the administered protective agents may be present in the same of different compositions.
  • the administered protective agents may be administered concurrently or sequentially.
  • the administered protective agents may be administered via the same or different routes of administration.
  • compositions and methods of the present invention are typically employed for the treatment of a mammal, preferably a human.
  • the acetaminophen intoxication includes both acute and chronic intoxication.
  • acute intoxication may be defined as consuming total > 150 mg/kg (about 7.5 g in adults) within 24 h. This equals 24 regular-strength tablets over 8 hours or less. However any other dose, including even one pill, may induce toxicity.
  • Chronic intoxication may occur as a result of long period of low dose consumption, excessive use or repeated overdose.
  • the symptoms of intoxication may include abdominal pain, appetite loss, coma, convulsions, diarrhea, irritability, jaundice, nausea, sweating, pallor, upset stomach and/or vomiting. Symptoms may also include elevations in the plasma levels of hepatic enzymes and bilirubin, prolongation of the prothrombin time, hypoglycemia and/or lactic acidosis.
  • administration of the compositions as disclosed herein provides a prolonged therapeutic window compared with the conventional, commercially available treatment, meaning that they are effective for a longer period of time following overdose or intoxication.
  • the therapeutic efficacy is maintained up to 10 hours or more, up to 10-15 hours, up to 15-24 hours, up to 24 hours or more, up to 48 hours or more.
  • the present invention further provides the use of the protective agents described herein, for the manufacture of a medicament for the treatment of drug intoxication.
  • the drug is acetaminophen.
  • mice Male C57BL/6, 9-10 or 11-12 weeks old
  • PC Phosphatidylcholine
  • Nutrasal Nutrasal.Com, Westbrook, ME 04092, made in USA
  • liquid kept in room temperature (R/T).
  • Concentration: 3000mg PC per 5ml 600mg/ml.
  • the solution was diluted to 60mg/ml (stock solution).
  • APAP N-acetyl-para-aminophenol
  • DDW di-distilized water
  • PBS phosphate buffer saline
  • mice were allocated into four groups of six mice each, as described in Table 1 below. The mice received sub-lethal hepatotoxic doses of APAP, causing liver damage equivalent to consumption of approximately 10 gr APAP (20 caplets of 500 mg) per 70kg man.
  • mice were treated according to the following procedure:
  • Table 2 presents the effect of each compound on the serum levels of the liver enzymes ALT and AST.
  • IB showed a protective effect when administered with APAP, leading to lower ALT/AST serum levels compared to administration of APAP alone (about 40% and 75% reduction, respectively, compared to administration of APAP only (p ⁇ 0.05).
  • this effect was observed for relatively low doses of IB, ranging from 0.1-0.4 mg/mouse.
  • mice that received APAP with IB showed lower body weight loss between Day 0 and Day 2, compared to mice that received APAP only (p ⁇ 0.01).
  • mice were allocated into four groups of six mice each, as detailed in Table 5 below. Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP, APAP+NAC or APAP+NAC+IB 15 hours later (9:00 next morning, Day 1) by gavage administration. Food was returned 2h after APAP administration (Day 1).
  • mice were weighted daily, and blood samples were taken 24h hours after APAP administration (Day 2) for liver enzyme tests.
  • liver biopsies were performed.
  • livers from individual mice were fixed in 10% formaldehyde solution and kept at room temperature until use.
  • the tissue blocks were then embedded in paraffin, 5-mm sections were cut and stained with hematoxylin and eosin (H&E) for morphological examination.
  • Figure 1 shows representative histological liver sections from mice taken 24h hours after APAP administration (X40 magnification).
  • diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis).
  • Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • Table 6 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • mice that received APAP with NAC+IB showed a significantly higher increase in body weight between Day 1 and Day 2, compared to mice that received APAP only (p ⁇ 0.05).
  • mice were allocated into four groups of six mice each, as described in Table 7 below. Mice were treated according to the following procedure:
  • mice were orally administered APAP, APAP+NAC or APAP+NAC+PC 15h hours later (9:00 next morning, Day 1) by gavage administration as follows: Group A: APAP
  • PC was administered 1.5 hours after APAP+NAC administration
  • mice were weighted daily, and blood samples were taken 24h hours after APAP administration (Day 2) for liver enzyme tests. In addition, liver biopsies were performed.
  • FIG. 2 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification).
  • APAP group diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • Table 8 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 9 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
  • mice that received APAP only showed a 6% decrease in their body weight during the two days of the experiment.
  • the combination of NAC+PC (administered after 1.5 or 3 hours) almost completely prevented body weight loss, and was significantly more effective than NAC alone (-99% versus -96%, p ⁇ 0.00001).
  • PC was administered 3 hours after APAP+NAC administration
  • FIG 3 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification).
  • APAP group diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • Table 11 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 12 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2 and by the difference in the mice body weight between Day 1 (immediately after administration of APAP with the different combinations and before food was returned) and Day 2.
  • mice that received APAP with NAC+PC 0.9 mg after 3 hours showed a significantly higher increase in body weight between Day 1 and Day 2, compared to mice that received APAP only (p ⁇ 0.01).
  • Administration of NAC alone did not show a significant effect.
  • PC was administered 3 hours after APAP+NAC administration
  • Table 14 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 15 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2 and by the difference in the mice body weight between Day 1 (immediately after administration of APAP with the different combinations and before food was returned) and Day 2.
  • MSM in combination with NAC was tested for its effect on toxicity induced by APAP using the same procedure described in Example 1 above.
  • Mice were allocated into three groups of six mice each, as detailed in Table 16 below. Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP, APAP+NAC or APAP+NAC+MSM 15 hours later (9:00 next morning, Day 1) by gavage administration. Food was returned 3.5h after APAP administration (Day 1).
  • Table 17 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 18 presents the effect of each combination on body weight, as determined by the difference in the mice body weight between Day 1 (immediately after administration of APAP with the different combinations and before food was returned) and Day 2.
  • Figure 4 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification).
  • APAP group diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • mice that received APAP with 3.5 mg NAC+MSM 1.5 mg showed a significantly higher increase in body weight between Day 1 and Day 2, compared to mice that received APAP only (p ⁇ 0.01).
  • the combination of NAC+MSM appeared to be more effective than NAC alone.
  • Example 6 Combination of NAC. MSM. PC and IB
  • mice were allocated into four groups of six mice each, as detailed in Table 19 below.
  • Food was taken on the afternoon (18:00) of Day 0.
  • Mice were orally administered APAP 15 hours later (9:00 next morning, Day 1) by gavage administration.
  • NAC was administered PO with APAP.
  • MSM+IB were administered 1.5h later.
  • PC was administered.
  • Food was returned 3.5h after APAP administration (Day 1). Weight was determined three times: on Day 0 before food was taken, after over-night fasting (Day 1, just before APAP administration) and before sacrifice (Day 2, 24 hours after APAP administration).
  • Table 20 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 21 presents the effect of each combination on body weight, as determined by the difference/ratio in the mice body weight between Day 1 and 2.
  • mice from each group were tested for their IFN- ⁇ secretion. After mice were sacrificed, spleens from five mice from each group were taken and splenocytes were isolated by the following procedure:
  • Spleens were stored in RPMI-1640 medium supplemented with FCS. Spleens were crushed through a 70- ⁇ nylon cell strainer (Falcon) and centrifuged (1,250 rpm for 7 min.). Red blood cells were lysed in 1 ml of cold 155 mM ammonium chloride lysis buffer. Splenocytes were washed and resuspended in 1 ml of RPMI supplemented with FCS. The viability of cells as assessed by trypan blue exclusion exceeded 90%.
  • splenocytes (lxlO 6 per mouse, per well, in duplicates) were moved into a 24 well plate containing RPMI-1640 supplemented with 10% FCS, 1% penicillin/streptomycin (P/S), 1 % glutamine, 1 % sodium pyruvate and 1% non-essential amino acid. All splenocytes were activated with 5 ⁇ g of Concanavalin A (purchased from MP Biomedicals, Ohio, USA), for 20 h at 37°C. After incubation cells from all wells were moved to Eppendorff tubes and centrifuged for 5 minutes. Then supernatants were collected and freezed in -80°C until assayed for IFN- ⁇ by ELISA.
  • Concanavalin A purchased from MP Biomedicals, Ohio, USA
  • Cytokine determination Levels of IFN- ⁇ were determined by "sandwich” ELISA using commercial kit according to the manufacturer's instructions (Quantikine, R&D Systems, Minneapolis, MN, USA).
  • Figure 5 shows IFN- ⁇ levels secreted by ConA-activated splenovytes derived from each group of mice.
  • the results have shown that oral administration of the above combination of MSM+IB with APAP is associated with a similar anti-inflammatory effect to that of APAP, while significantly alleviating the APAP-induced liver injury.
  • mice treated with lOmg of APAP alone or in combination with 0.1 mg IB + 0.25 mg MSM were followed for APAP serum levels.
  • Serum levels of APAP in mice treated with 10 mg of APAP alone were similar to mice treated with APAP+IB+MSM (129 vs. 149 ug/ml, respectively. p NS).
  • mice were allocated into four groups of six mice each, as described in Table 26 below.
  • the mice received sub-lethal hepatotoxic doses of APAP (4.6mg/mouse).
  • the effect of the different combinations of compounds on toxicity induced by APAP was tested using the procedure described in Example 1 above.
  • Figure 6 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification).
  • APAP group diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • Table 27 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 30 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
  • **PC was administered 3 hours after APAP+NAC or after APAP+Cys or after APAP+ Met; were administered.
  • mice were treated according to the following procedure:
  • Figure 7 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification).
  • APAP group diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • Table 30 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
  • Table 33 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
  • Figure 8 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification).
  • APAP group diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
  • Table 33 presents the effect of various treatments on the serum levels of the liver enzymes ALT and AST.
  • Table 36 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
  • APAP with NAC resulted in about 85-87% reduction in the serum levels of ALT and AST compared to administration of APAP alone.
  • Administration of APAP with Cys or Met resulted in a greater reduction in the liver enzymes, up to about 94-98% reduction compared to APAP alone (p ⁇ 0.0005).
  • mice that received APAP only showed more than 6% decrease in their body weight during the two days of the experiment.
  • the combination of APAP with NAC resulted in lower weight loss, to 98.08%.
  • NAC was replaced by either Cys or Met, the effect on body weight was similar (97-99% p ⁇ 0.007).

Abstract

Compositions comprising isosilybin B and optionally additional protective agents selected from methylsulfonylmethane (MSM), phosphatidylcholine (PC), cysteine (Cys), seleno-cysteine (Se-Cys), ribose-cysteine (RibCys), N-acetylcysteine (NAC), N- acetylcysteine-amide (AD4), methionine (Met) and S-adenosylmethionine (SAM) for use in reducing and / or preventing drug- induced toxicity, such as acetaminophen-induced toxicity. Said compositions further comprising acetaminophen for use as safe formulations of acetaminophen with reduced risk of causing liver damage.

Description

COMPOSITIONS AND METHODS FOR AMELIORATION AND PREVENTION
OF DRUG-INDUCED TOXICITY
FIELD OF THE INVENTION
The present invention relates to the treatment and prevention of drug-induced toxicity, such as liver and kidney toxicity. In particular, the present invention relates to compositions and methods useful in treating, reducing and/or preventing liver failure, liver injury, liver damage and other toxicity effects associated with acetaminophen or caused by acetaminophen overdose.
BACKGROUND OF THE INVENTION
Acetaminophen (chemically named N-(4-hydroxyphenyl)acetamide), also known as para-acetylaminophenol, paracetamol and N-acetyl-para-aminophenol (abbreviated as APAP), is a widely used analgesic and antipyretic drug. It is commonly used for the relief of headaches and other minor pains, and is a major ingredient in various cold and flu remedies.
Acetaminophen is the generic name of a drug found in many common brand-name over-the-counter (OTC) and prescription (Rx) products. In some products acetaminophen is the sole active ingredient (e.g., TYLENOL®), while in others it is combined with additional active ingredients, such as opioid analgesics, nasal decongestants, cough suppressants and antihistamines. Examples include combinations with oxycodone (e.g. PERCOCET®) and hydrocodone (e.g. VICODIN®). Acetaminophen is available as a tablet, capsule, liquid suspension, suppository, intravenous, and intramuscular form.
While generally safe for use at recommended doses (up to 1,000 mg per single dose and up to 4,000 mg per day for adults), acute overdoses of acetaminophen can cause serious liver injury, acute liver failure (ALF) and even death. In effect, acetaminophen- induced liver toxicity is the most prevalent cause of acute liver failure in the Western world. In addition, several drug-host interactions, such as drug viral interactions were suggested as potential causes for acetaminophen-induced liver toxicity even without exceeding the recommended dosages.
In the liver, acetaminophen is metabolized through several pathways before being eliminated from the body. One of these pathways, which involves the cytochrome P-450 systems, yields a toxic intermediate known as N-acetyl-p-benzo-quinone imine (NAPQI). Normally, this toxic metabolite is detoxified by conjugation to reduced glutathione. However, an excessive amount of acetaminophen in the liver, and accordingly excessive amounts of NAPQI, results in shortage of reduced glutathione and accumulation of the toxic metabolite. The toxic metabolite then binds to liver proteins and cause cellular injury. The amount of the accumulated toxic metabolite and the difficulty of the liver to eliminate it before it causes damage influence the extent of liver injury. NAPQI accumulation is only one of the suggested explanations for acetaminophen-induced liver toxicity.
In addition, the immune system was suggested to play a major role in the pathogenesis of the toxic effect and the liver injury. Both NK and NKT cells as well as several cytokines were suggested as important factors in this context.
Despite a number of efforts since the early 1990s to reduce the incidence of acetaminophen-related liver injury, the extent of liver failure cases reported in the medical literature indicates that liver injury from acetaminophen overdose remains a serious public health problem.
In addition to the liver, renal damage is also associated with acetaminophen consumption. In toxic amounts, acetaminophen may result in acute tubular necrosis. At therapeutic dosages, acetaminophen can be toxic in patients with risk factors such as starvation and chronic ingestion of alcohol or certain medications. Long term consumption of normal doses of acetaminophen has also been found to result in cumulative damage to the kidneys.
Many cases of acetaminophen overdose are caused unintentionally by consumers inadvertently taking more than the recommended dose. A number of factors that may contribute to acetaminophen overdose problem have been identified, including: the narrow safety margin of acetaminophen, risk factors rendering some individuals specially and specifically prone to acetaminophen-induced liver injury, such as excessive alcohol consumption and pre-existing liver disorders, insufficient public knowledge about the serious risks of acetaminophen overdose, and the availability of many different types of OTC and Rx products containing acetaminophen and possibly other active ingredients, and a variety of doses for different indications. The latter may result in the consumption of more than one acetaminophen containing product without realizing that acetaminophen is included in all of these products, and accordingly an increased risk of acetaminophen overdose. In addition, it can be difficult to recognize and diagnose the onset of liver injury. Current treatments of acetaminophen overdose are mainly aimed at removing acetaminophen from the body and trying to replace diminished glutathione levels. Oral administration of activated charcoal can also be used to decrease absorption of acetaminophen by binding any drug remaining in the gastrointestinal tract. The treatment also includes administration of an antidote, N-acetylcysteine (NAC), which is a known precursor for glutathione, thereby helping the body to regenerate reduced glutathione and prevent or reduce damage to the liver. A liver transplant is sometimes required if damage to the liver becomes severe and irreversible.
The known treatments have several drawbacks, mainly due to their limited therapeutic window. Actions such as gastric lavage or administration of activated charcoal are effective only in cases where the patients present for treatment soon after taking the overdose. NAC is considered effective only when administered within 8 hours following intoxication, after which the efficacy is significantly reduced.
Patients with acute overdose, or with liver toxicity due to any dose, can deteriorate into fulminant liver failure. Although some of these patients will recover spontaneously, for some of them, liver transplantation is the only way to prevent deterioration and death.
There are currently no commercially available formulations of acetaminophen which purposely intended to reduce the risk of liver toxicity.
US 4,868,114 discloses a method comprising stimulating the biosynthesis of glutathione in mammalian cells by contacting the cells with an effective amount of cysteine prodrugs.
US 6,555,141 discloses therapeutic compositions for the protection, treatment and repair of liver tissue comprising two or more compounds selected from the group consisting of S-adenosylmethionine, L-ergothioneine, and a compound selected from the group consisting of Milk thistle (Silybum marianum), silymarin and active components of silymarin, whether naturally, synthetically, or semi-synthetically derived, and to methods of preventing and treating liver disease and of repairing damaged liver tissue.
US 6,566,401 discloses pharmaceutical compositions for the treatment or prevention of the toxic effects of therapeutic agents and methods of treating or preventing such toxicity using a toxicity reducing amount of N-acetylcysteine either alone or in combination with a therapeutically effective amount or, to achieve its therapeutic advantages, an amount larger than what is customarily given as a therapeutically effective amount, of a therapeutic agent. US 7,238,373 discloses a nutritional supplement that is designed to provide nutritional benefits as well as to assist the body with detoxification.
US 8,148,356 discloses acetylcysteine compositions in solution, comprising acetylcysteine and which are substantially free of metal chelating agents, such as EDTA. Further, this invention relates to methods of making and using the acetylcysteine compositions. The present compositions and methods are designed to improve patient tolerance and compliance, while at the same time maintaining the stability of the pharmaceutical formulation. The compositions and methods of this invention are useful in the treatment of acetaminophen overdose, acute liver failure, various cancers, methacrylonitrile poisoning, reperfusion injury during cardio bypass surgery, and radiocontrast-induced nephropathy, and can also be used as a mucolytic agent.
US 2011/0124718 discloses the reduction of acetaminophen toxicity by dietary milk thistle extract. Milk thistle and acetaminophen are combined to provide single dosages containing both ingredients, whether dispensed in a liquid suspension, chewable tablets, coated caplets, gelcaps, geltabs, and suppositories.
US 2012/0022161 discloses a pharmaceutical composition and method for providing a reduction in side effects for human patients in need of therapy comprising the administration of a pharmaceutical composition comprising acetylcysteine.
WO 2009/097874 discloses compositions comprising a combination of amino acids selected from: a) L-Cysteine with L-Methionine; b) L-Cysteine with L-Serine; c) L- Cysteine with L-Methionine and L-Serine; d) L-Methionine with L-Serine; or precursors or transporters of said amino acids, for the prevention of paracetamol-induced liver damage.
WO 2011/044230 discloses methods and compositions comprising N- acetylcysteine amide (NAC amide) and derivatives thereof for use in treatments and prophylactic therapies for human and non-human mammalian diseases, disorders, conditions and pathologies associated with bomb blast or other high energy noise or impulse blasts. Pharmaceutically or physiologically acceptable compositions of NAC amide or derivatives thereof are also provided, which may be administered alone, or in combination with other suitable agents.
Polyak et al. (2010) PNAS U.S.A, 107; (5995 - 5999), report the identification of hepatoprotective flavonolignans from silymarin. There still remains an unmet need for more effective compositions and methods for treating or preventing drug toxicity, e.g. liver damage caused by acetaminophen. There is a further need for novel compositions of acetaminophen characterized by decreased risk of inducing toxicity while preserving efficacy. Such compositions may be useful for reducing or even preventing acetaminophen-related toxicity.
SUMMARY OF THE INVENTION
The present invention provides pharmaceutical compositions comprising combinations of protective agents which are effective in reducing and/or preventing drug- induced toxicity, including but not limited to, acetaminophen-induced toxicity. The protective agents utilized herein are selected from isosilybin B, methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno-cysteine (Se-Cys), ribose-cysteine (RibCys), N-acetylcysteine (NAC), N-acetylcysteine-amide (AD4), methionine (Met) and S-adenosylmethionine (SAM).
The pharmaceutical compositions containing the protective agents may be formulated with or without a drug or substance with potential toxicity, and accordingly may be used as safe formulations of the drug, or as an antidote in case of intoxication.
According to certain embodiments, the compositions are formulated with or without acetaminophen. Acetaminophen-containing compositions may be used as safe formulations with reduced risk of causing liver damage and/or renal damage. Compositions without acetaminophen can be used as an antidote for the treatment of acetaminophen-related toxicity, such as liver failure or liver disorder.
The present invention further provides methods for treating drug-induced intoxication, such as acetaminophen-induced intoxication, utilizing the combinations of protective agents disclosed herein.
The combinations of protective agents of the present invention were found to be remarkably effective in reducing liver damage and liver toxicity associated with acetaminophen administration. As exemplified herein below, mice exposed to sub-lethal amounts of acetaminophen administered with different combinations of the above compounds showed significantly reduced serum levels of liver enzymes and better body weight recovery following fast compared to mice exposed to same amounts of acetaminophen alone. In addition, histological examination revealed little or no liver damage in mice that received the different combinations of protective compounds, in contrast to mice who received acetaminophen alone. The ability of the compounds to protect the liver was demonstrated when the compounds and acetaminophen were administered concurrently, as well as when the compounds were administered several hours following acetaminophen administration. The combinations of the present invention act at least additively and preferably synergistically and are superior to the currently used antidote (gold standard treatment) of acetaminophen poisoning, namely, NAC alone.
According to one aspect, the present invention provides a pharmaceutical composition comprising isolated isosilybin B, and optionally further comprising at least one additional protective agent selected from the group consisting of methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno-cysteine (Se- Cys), ribose-cysteine (RibCys), N-acetylcysteine (NAC), N-acetylcysteine-amide (AD4), methionine (Met) and S-adenosylmethionine (SAM), wherein said pharmaceutical composition is effective in treating or preventing drug-induced toxicity.
As will be explained in more detail below, isosilybin B is one of the components of an extract produced from milk thistle (Silybum marianum). This compound is utilized herein in an isolated form, meaning that it is substantially separated from other components of the extract.
In some embodiments of the present invention, the composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
In some embodiments of the present invention, the pharmaceutical composition further comprises a pharmaceutically acceptable diluent, excipient or carrier.
In some embodiments of the present invention, the phosphatidylcholine component comprises a majority of unsaturated fatty acids. In some embodiments, the phosphatidylcholine component comprises a majority of polyunsaturated fatty acids. In some embodiments, the polyunsaturated phosphatidylcholine comprises unsaturated fatty acids selected from the group consisting of linoleic acid, linolenic acid and oleic acid. In some embodiments, linoleic acid constitutes about 70% or more of the fatty acid content of the phosphatidylcholine.
In some embodiments of the present invention, the phosphatidylcholine component comprises 1, 2-dilinoleoylphosphatidylcholine (DLPC), with linoleic acid bound both in cl- and c2-positions of the molecule. In some embodiments, about 50% or more of the phosphatidylcholine molecules present within the composition are DLPC.
In some embodiments of the present invention, the pharmaceutical composition is used for the treatment of acetaminophen intoxication.
In some embodiments of the present invention, the protective agents described herein are formulated together with a drug that can potentially cause toxic effects, in a single pharmaceutical composition. Thus, in some embodiments, the pharmaceutical composition of the present invention further comprises a drug with potential toxicity. In some embodiments, the drug is acetaminophen. According to these embodiments, the pharmaceutical composition is characterized by a reduced risk of acetaminophen-induced toxicity, such as acetaminophen-induced liver damage and/or nephrotoxicity, compared to formulations of acetaminophen that do not contain the protective agents as described herein. Such compositions are contemplated to substantially retain the efficacy of the drug, e.g. acetaminophen, while significantly reducing or preventing drug-induced liver toxicity, liver damage and kidney damage.
In additional embodiments, the acetaminophen-containing pharmaceutical composition of the present invention further comprises at least one additional drug selected from the group consisting of an opioid analgesic, a decongestant, a cough suppressant, an antihistamine and an expectorant.
In some embodiments of the present invention, the opioid analgesic is selected from the group consisting of codeine, oxycodone, hydrocodone and propoxyphene.
In some embodiments, the decongestant is selected from the group consisting of phenylephrine and pseudoephedrine.
In some embodiments of the present invention, the expectorant is guaiphenesin.
In some embodiments of the present invention, the cough suppressant is selected from the group consisting of dextromethorphan and codeine.
In some embodiments of the present invention, the antihistamine is selected from the group consisting of chlorpheniramine, phenyltoloxamine and doxylamine.
Typically, the pharmaceutical composition is formulated for systemic administration. In some embodiments, the pharmaceutical composition is formulated for a route of administration selected from the group consisting of oral, rectal, intravenous, intramuscular, intradermal, subcutaneous and intranasal. According to another aspect, the present invention provides a method for treating drug intoxication in a subject in need thereof, the method comprising administering to the subject isolated isosilybin B, and optionally at least one additional protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys) Se-Cys, RibCys, NAC, AD4, Met and SAM.
In some embodiments of the present invention, the method comprises administering to the subject isolated isosilybin B and further administering (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
When two or more protective agents are used to treat the drug intoxication, in some embodiments of the present invention, the protective agents are within a single composition. In other embodiments of the present invention, the protective agents are within separate compositions. In some embodiments, the two or more protective agents are administered concurrently. In other embodiments, the two or more protective agents are administered sequentially. In some embodiments, the two or more protective agents are administered via the same route of administration. In other embodiments, the two or more protective agents are administered via different routes of administration.
In some embodiments of the present invention, administration is performed via a route of administration selected from the group consisting of oral, rectal, intravenous, intramuscular, intradermal, subcutaneous and intranasal. In some embodiments, the route of administration is selected from the group consisting of oral and intravenous.
In some embodiments, the method of the present invention is applied to a subject who has consumed, or suspected of having consumed, large amounts of acetaminophen. In some embodiment, acetaminophen intoxication is acute intoxication. In other embodiments, acetaminophen intoxication is chronic intoxication.
In some embodiments of the present invention, the subject is human. In other embodiments, the subject is a non-human mammal.
According to another aspect, the present invention provides a composition comprising isolated isosilybin B, and optionally at least one additional protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, for use in the treatment of drug intoxication.
In some embodiments, the drug is acetaminophen. In some embodiments of the present invention, the pharmaceutical composition further comprises a pharmaceutically acceptable diluent, excipient or carrier.
In some embodiments of the present invention, the pharmaceutical composition is formulated for administration via a route of administration selected from the group consisting of oral, rectal, intravenous and intramuscular.
These and further aspects and features of the present invention will become apparent from the detailed description, examples and claims which follow.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1. Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus IB, compared to mice administered with acetaminophen only.
Figure 2. Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus PC, compared to mice administered with acetaminophen only.
Figure 3. Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus PC, compared to mice administered with acetaminophen only.
Figure 4. Representative liver biopsy results of mice treated with acetaminophen in combination with NAC alone or NAC plus MSM, compared to mice administered with acetaminophen only.
Figure 5. IFN-γ levels secreted by ConA-activated splenocytes derived from acetaminophen-intoxicated mice.
Figure 6. Representative liver biopsy results of mice treated with acetaminophen, acetaminophen plus NAC, acetaminophen plus Cys, PC, MSM and IB, or acetaminophen plus Met, PC, MSM and IB.
Figure 7. Representative liver biopsy results of mice treated with acetaminophen, acetaminophen plus NAC, PC, MSM and IB, acetaminophen plus Cys, PC, MSM and IB, or acetaminophen plus Met, PC, MSM and IB.
Figure 8. Representative liver biopsy results of mice treated with acetaminophen, acetaminophen plus NAC, acetaminophen plus Cys, or acetaminophen plus Met. DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to treatment and prevention of drug-induced toxicity, particularly acetaminophen-induced toxicity, using protective active ingredients selected from the group consisting of isosilybin B, methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno-cysteine (Se-Cys), ribose-cysteine (RibCys), N- acetylcysteine (NAC), N-acetylcysteine-amide (AD4), methionine (Met) and S- adenosylmethionine (SAM). Typically, a plurality of protective active compounds is used.
As used herein, "a plurality" indicates at least two.
As used herein, a "protective agent" or "protective compound" refers to a compound effective in treating or preventing drug-induced toxicity. In particular, the term refers to a compound that serves to decrease hepatotoxicity and/or nephrotoxicity caused by the consumption of a drug. The term "drug" is used herein to refer to a compound with a therapeutic activity, most often a manmade small organic molecule, that can potentially cause toxic effects, such as damage to the liver and possible other organs, for example, acetaminophen. For the purposes of the application and claims, a drug is a molecule known to exhibit potential hepatotoxicity.
As used herein, the phrase "pharmaceutical composition effective in treating or preventing drug-induced toxicity", indicates that the protective agents are present in the pharmaceutical composition in an amount that is effective to perform their utility. When a drug is included in the composition, the phrase indicates that the protective agents are present in the composition in an amount that is effective to significantly reduce or even completely inhibit toxic effects of the drug. When a drug is not included in the composition, the phrase indicates that the amount of protective agents is effective in alleviating or even eliminating liver injury and/or other damage caused by the consumption of a drug.
As used herein "drug-induced toxicity" refers to damages to body tissues and organs resulting from the consumption of a drug, typically consumption of large amounts of the drug.
As used herein, the term "potential toxicity", when referring to a drug, indicates that the drug is generally safe when consumed at the recommended doses, but can be toxic when large amounts are consumed, and/or when consumed by a subject who is more susceptible to toxic effects of the drug. As used herein, "drug intoxication" refers to a clinical condition resulting from the consumption of toxic amounts of a drug (e.g. acetaminophen) that damage the liver and/or other organs such as the kidneys.
As used herein, the terms "treating" and "treatment", when referring to drug- induced toxicity or drug intoxication, encompass reducing or even eliminating the drug- induced (for example, acetaminophen-induced) liver damage, and/or renal damage. The terms may also encompass improvement of other symptoms associated with intoxication, as detailed hereinbelow. The extent of liver damage, as well as improvement and reduction in liver damage as a result of utilizing the compositions and methods of the present invention, may be evaluated, for example, by measuring serum levels of liver enzymes. An increased level of liver enzymes in the serum indicates liver damage. Nephrotoxicity can be monitored, for example, through simple blood and urine tests. A decreased glomerular filtration rate (GFR) indicates renal damage and poor renal function.
As used herein, the term "preventing", when referring to drug-induced toxicity, encompasses significantly reducing the risk of toxicity or even completely inhibiting toxic effects of the drug.
As used herein, the term "about", when referring to a measurable value such as an amount, is meant to encompass variations of +/-10%, more preferably +1-5%, even more preferably +/-1 %, and still more preferably +/-0.1 % from the specified value, as such variations are appropriate to achieve the intended purpose.
Pharmaceutical compositions
Pharmaceutical compositions of the present invention comprise protective agents selected from isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM. Pharmaceutically acceptable salts of the protective agents described herein are also within the scope of the present invention. The compositions may be formulated with or without a drug with potential toxicity, such as acetaminophen.
According to an aspect of the present invention, there is provided herein a pharmaceutical composition effective in treating or preventing drug-induced, such as acetaminophen-induced, liver toxicity, the composition comprising isolated isosilybin B , and optionally further comprising at least one more protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM. Each combination represents a separate embodiment of the invention. According to another aspect of the present invention, there is provided herein a pharmaceutical composition comprising the combinations of protective agents disclosed herein, for use in the treatment of drug intoxication. In particular embodiments, the drug is acetaminophen.
In some embodiments, the pharmaceutical composition comprises isosilybin B. In some embodiments, isosilybin B is combined with at least one more protective agent selected from the group consisting of NAC, phosphatidylcholine, MSM, RibCys, AD4, Cys, Se-Cys, Met and SAM.
In some embodiments, the pharmaceutical composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
In some embodiments, the pharmaceutical composition comprises isolated isosilybin B, NAC and one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
In other embodiments, the pharmaceutical composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
In some embodiments, the pharmaceutical composition comprises isolated isosilybin B and further comprises (i) one or more protective agents selected from the group consisting of Cys and Met; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
In some embodiments, a pharmaceutical composition effective in treating or preventing drug-induced toxicity is provided, the composition comprises a plurality of protective agents selected from the group consisting of isolated isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM. Each combination represents a separate embodiment of the invention.
In some embodiments, the composition comprises NAC and at least one more protective agent selected from the group consisting of phosphatidylcholine, MSM, isosilybin B. In some embodiments, the combination of NAC and at least one more protective agent selected from the above compounds permits lower amounts of NAC in order to achieve a therapeutic effect compared to the amount required for NAC alone. In some embodiments, the composition comprises (i) one or more protective agents selected from the group consisting of AD4, Cys, Se-Cys, RibCys, Met and SAM; and (ii) one or more protective agents selected from the group consisting of phosphatidylcholine, MSM, isosilybin B.
In some embodiments, the composition comprises a combination of two or more protective agents selected from the group consisting of NAC, phosphatidylcholine, MSM, isosilybin B, RibCys and SAM.
In some embodiments, the pharmaceutical composition comprises NAC and phosphatidylcholine as the protective agents. In additional embodiments, the pharmaceutical composition comprises NAC in combination with MSM or SAM as the protective agents. In yet additional embodiments, the pharmaceutical composition comprises NAC and isosilybin B as the protective agents. In some embodiments, the combination of NAC and at least one more protective agent selected from the above compounds permits lower amounts of NAC in order to achieve a therapeutic effect compared to the amount required for NAC alone.
In some embodiments, the pharmaceutical composition does not include NAC.
In some embodiments of the present invention, the compositions contain the protective agents described herein, and do not contain a drug that can cause intoxication. Such compositions are effective in treating drug-induced toxicity, and can be administered in case that drug-induced toxicity has occurred, in order to treat said toxicity. For example, such compositions can be administered following an overdose of a drug, which results in signs of intoxication, in order to treat the toxicity effects. In particular embodiments, the pharmaceutical compositions of the present invention can be used as an antidote for the treatment of acetaminophen intoxication.
In other embodiments, the compositions of the present invention contain the protective agents described herein, and further contain a drug that can cause intoxication. Such compositions are characterized by a reduced risk of drug intoxication compared to compositions that contain the drug without the protective agents.
In particular embodiments, the compositions contain acetaminophen and possibly additional one or more drugs. According to these embodiments, safe formulations of acetaminophen are provided, with significantly reduced risk of causing liver and renal damage, even when taken at large amounts. Such compositions have several advantages, including a significantly reduced risk for overdose and significantly reduced need for liver transplantation following an overdose. In addition, such compositions may be beneficial to populations at risk.
In some embodiments, a pharmaceutical composition is provided, which comprises acetaminophen, isosilybin B and at least one more protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, said pharmaceutical composition is characterized by a reduced risk of drug intoxication compared to a pharmaceutical composition comprising acetaminophen without isosilybin B and the at least one more protective agent.
In some embodiments, a pharmaceutical composition is provided, which comprises acetaminophen and further comprises a plurality of protective agents selected from the group consisting of isolated isosilybin B, MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM, said pharmaceutical composition is characterized by a reduced risk of drug intoxication compared to a pharmaceutical composition comprising acetaminophen without the plurality of protective agents.
The protective agents utilized herein are commercially available and may also be synthesized or extracted by methods known in the art or by new methods. NAC may be identified by CAS Registry Number 616-91-1, MSM may be identified by CAS Registry Number 67-71-0, isosilybin B may be identified by CAS Registry Number 142796-22-3, SAM may be identified by CAS Registry Number 29908-03-0, RibCys may be identified by CAS Registry Number 17087-36-4, AD4 may be identified by CAS Registry Number 38520-57-9, Cys may be identified by CAS Registry Number 52-90-4, Se-Cys may be identified by CAS Registry Number 10236-58-5, Met may be identified by CAS Registry Number 63-68-3.
Phosphatidylcholine is a phospholipid composed of a choline headgroup and glycerophosphoric acid with a variety of fatty acids. The fatty acids may include saturated and unsaturated fatty acids.
In some embodiments of the present invention, the phosphatidylcholine component comprises mostly unsaturated fatty acids. In some embodiments, the fatty acid content of the phosphatidylcholine comprises at least one of linoleic acid, linolenic acid and oleic acid. In some embodiments, linoleic acid constitutes about 70% or more of the fatty acid content of the phosphatidylcholine.
In some embodiments of the present invention, the phosphatidylcholine component comprises 1, 2-dilinoleoylphosphatidylcholine (DLPC), with two linoleic acid molecules. In some embodiments, about 50% or more of the phosphatidylcholine molecules present within the composition are DLPC.
Isosilybin B (also known as isosilibinin B) is a flavonolignan that constitutes one of the components of the milk thistle (Silybum marianum) extract. Traditional milk thistle extract is made from the plant seeds and consists of about 65-80% silymarin (a flavonolignan complex) and 20-35% fatty acids. Silymarin is a complex mixture of polyphenolic molecules, including seven closely related flavonolignans, namely silybin A, silybin B, isosilybin A, isosilybin B, silychristin, isosilychristin and silydianin, and one flavonoid (taxifolin).
It is now disclosed that isosilybin B is highly more potent and effective in reducing acetaminophen-induced liver toxicity, compared to other compounds found in silymarin, or in any other milk thistle extract of any part of the plant. As exemplified herein below, the closely related silybin B and isosilybin A did not show a beneficial effect when administered to mice together with, or following, large dosage of acetaminophen. Isosilybin B may be purified from an extract of the milk thistle. Isosilybin B is utilized herein in an isolated form, that is separated from the other ingredients of the extract. As used herein, "isolated" indicates at least about 80%, separated from other ingredients of the extract, preferably at least about 90%, more preferably at least about 95% separated from other ingredients of the extract. Synthetic Isosilybin B may also be used.
In some embodiments, the NAC is substantially free of its oxidized form, di-n- acetylcysteine. It is preferred that the therapeutic agent, serially or co-administered, be in any form in which it is typically available and the composition should be prepared in a manner that substantially prevents oxidation of the NAC during preparation or storage.
In some embodiments of the invention, the preparation and storage of the formulation is performed in such a way that the reduced form of NAC is the primary form administered to the patient. Maintaining NAC containing formulations in solid form is preferable for this purpose. When in solution, NAC containing formulations are preferably stored in a dark bottle that is vacuum sealed. Storage in cool dark environments is also preferred.
The determination of reduced and oxidized species present in a sample may be determined by various methods known in the art, for example with capillary electrophoresis, HPLC, etc. as described by Chassaing et al. (1999) J Chromatogr B Biomed Sci Appl 735(2):219-27. The protective agents described herein are present in the composition in an amount which is effective for inducing the therapeutic effect.
Identification of therapeutically effective amounts can be performed by methods well known to those skilled in the art. Exemplary dosage ranges for the different active protective agents (for human use, oral administration) include doses ranging between about 0.05 - 10 mg/kg, for example between about 0.05-5 mg/kg, between about 0.05-1 mg/kg, between about 0.1 - 2.5 mg/kg, between about 0.5-2 mg/kg.
In some embodiments, when two or more protective agents are present in the composition, they can be formulated such that they are released concurrently. In other embodiments, they are formulated such that they are released sequentially. For example, in case there are two protective agents within a pharmaceutical composition, one can be formulated for immediate release, and the other for sustained release. Such formulations are known in the art.
In some embodiments, the protective agents are formulated within liposomes. The liposomes may be used as a delivery method. The liposomes may be used for liver targeting. One or more of the protective agents may be used to create the liposomes.
In some embodiments, the pharmaceutical composition is a combination medication that further comprises acetaminophen. The composition may also include additional one or more drugs, such opioid analgesics, decongestants, cough suppressants, antihistamines, expectorants or combinations thereof. Information about combination medications containing acetaminophen and at least one more active ingredient can be found, for example, at the MedLinePlus website (www.nlm.nih.gov/medlineplus/).
Non-limiting examples of opioid analgesics include codeine, oxycodone, hydrocodone and propoxyphene. Non-limiting examples of decongestants include phenylephrine, pseudoephedrine and guaiphenesin. Non-limiting examples of cough suppressants include dextromethorphan and codeine. Non-limiting examples of antihistamines include chlorphenir amine, phenyltoloxamine and doxylamine. A non- limiting example of an expectorant is guaiphenesin.
Advantageously, the combination of the protective compounds described herein with acetaminophen (and optionally other drugs) does not interfere with the normal pharmaceutical activity of acetaminophen, and it maintains its pain relieving and fever reducing properties. Acetaminophen is commercially available and may also be synthesized by methods well known in the art. An overview of synthetic processes of acetaminophen can be found, for example, in Anthony S. Travis (2007). "Manufacture and uses of the anilines: A vast array of processes and products". In Zvi Rappoport. The chemistry of Anilines Part 1. Wiley, p. 764; and Elmar Friderichs, Thomas Christoph, Helmut Buschmann (2005), "Analgesics and Antipyretics", Ullmann's Encyclopedia of Industrial Chemistry, Weinheim: Wiley- VCH
The compositions of the present invention that comprise acetaminophen may find use as anti-pyretic and analgesic agents, and are suitable for medical indications treatable with acetaminophen alone. An improvement in product safety is provided by the inclusion of the protective compounds as disclosed herein, which substantially prevents the possibility of accidental or inadvertent overdose.
The amount of compounds in the compositions of the present invention which will be effective in the treatment of a particular condition will depend on the nature of the condition, and can be determined by standard clinical techniques. See, for example, Goodman and Oilman; The Physician's Desk Reference, Medical Economics Company, Inc., Oradell, N.J., 1995; and to Drug Facts and Comparisons, Facts and Comparisons, Inc., St. Louis, Mo., 1993.
As the addition of the compounds as disclosed herein does not affect the therapeutic efficacy of acetaminophen, it is generally not necessary to adjust the dosage from what would ordinarily be administered for acetaminophen alone, and in fact the dose may be raised due to the increased safety of the present formulations. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease, and should be decided according to the judgment of the practitioner and each patient's circumstances.
For example, the acetaminophen present in orally administrable solid unit doses will usually be at least about 80 mg (for pediatric doses), 325 mg, 500 mg and 650 mg. Oral liquid dosage forms usually comprise at least about 100 mg/ml, 120 mg/2.5 ml, 120mg/5 ml, 160 mg/5 ml, 165 mg/5 ml, 325 mg/5 ml acetaminophen.
Suppositories are formulated in the manner well known in the art and usually comprise at least about 120 mg, 125 mg, 325 mg, 500 mg and 650 mg acetaminophen per dosage unit Typically, the composition further comprises a pharmaceutically acceptable diluent, excipient or carrier.
As used herein, the term "pharmaceutically acceptable diluent, excipient, or carrier" refers to a diluent, excipient, or carrier that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered active agent.
As used herein, the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Non-limiting examples of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols. Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences", Mack Publishing Co., Easton, PA, (Remington: The Science and Practice of Pharmacy, Gennaro, A., Lippincott, Williams & Wilkins, Philadelphia, Pa., 20th ed, 2000).
As used herein, a "carrier" refers to any substance suitable as a vehicle for delivering of the agents or molecule of the present invention to a suitable in vivo or in vitro site. As such, carriers can act as a pharmaceutically acceptable excipient of a therapeutic composition of the present invention. Carriers of the present invention include: (1) excipients or formularies that transport, but do not specifically target a molecule to a cell (referred to herein as non-targeting carriers); and (2) excipients or formularies that deliver a molecule to a specific site in a subject or a specific cell (i.e., targeting carriers). Examples of non-targeting carriers include, but are not limited to water, phosphate buffered saline, Ringer's solution, dextrose solution, serum-containing solutions, Hank's solution, other aqueous physiologically balanced solutions, oils, esters and glycols. Aqueous carriers can contain suitable auxiliary substances required to approximate the physiological conditions of the recipient, for example, by enhancing chemical stability and isotonicity.
Therapeutic compositions of the present invention can be sterilized by conventional methods.
Any suitable route of administration may be used for the composition of the present invention, including but not limited to local and systemic routes. Systemic administration includes all enteral and all parenteral routes. Non-limiting examples of suitable administration routes include oral, rectal, transmucosal such as transnasal and buccal, intravenous, intramuscular, transdermal, subcutaneous, intradermal, intravitreal, intravesicular and inhalation routes.
Thus, in some embodiments, the composition of the present invention is formulated for topical administration. In other embodiments, the composition is formulated for systemic administration.
Pharmaceutical compositions of the present invention may be formulated in conventional manners. The proper formulation is dependent upon the route of administration chosen.
In some embodiments, the compositions of the present invention are formulated for oral administration. Non-limiting examples of formulations for oral administration include tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Suitable carriers for oral administration are well known in the art. Compositions for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries as desired, to obtain tablets or dragee cores. Non-limiting examples of suitable excipients include fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol, cellulose preparations such as, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, and sodium carbomethylcellulose, and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate, may be added.
For administration by injection, the active ingredients of the composition may be formulated in aqueous solutions, for example in physiologically compatible buffers including but not limited to Hank's solution, Ringer's solution, or physiological salt buffer. Formulations for injection may be presented in unit dosage forms, for example, in ampoules, or in multi-dose containers with, optionally, an added preservative. The compositions may be in the form of suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing, and/or dispersing agents. Non-limiting examples of suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters such as ethyl oleate, triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the active ingredients, to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, for example, a sterile, pyrogen-free, water-based solution, before use. For intravenous administration, it is preferable to adjust the pH of the administered solution to that of the blood, namely, about pH=7.4.
For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by inhalation route, the active ingredients are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluorome thane, dichloro-tetrafluoroethane, or carbon dioxide. In the case of a pressurized aerosol, the dosage may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, for example, gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base, such as lactose or starch.
In some embodiments, the compositions of the present invention are formulated for rectal administration, for example, as suppositories or retention enemas, using, for example, conventional suppository bases such as cocoa butter or other glycerides.
The exact formulation, route of administration, and dosage can be chosen by the individual physician in view of the patient's condition.
In some embodiments, the composition further comprises at least one additive useful in the pharmaceutical fields, including, but not limited to fats, emulsifiers and co- emulsifiers, hydrophilic or lipophilic gelling agents, colorants, fragrances, emollients, humectants, preservatives, vitamins, chelators, solvents, fillers, thickeners, hydrophilic and lipophilic filters, dyestuffs, neutralizers, penetration-enhancing agents and polymers.
Non-limiting examples of suitable fats include mineral oils, oils of animal origin (lanolin), synthetic oils (isopropyl myristate, octyldodecyl, isostearyl isostearate, decyl oleate or isopropyl palmitate), silicone oils (cyclomethicone or dimethicone) and fluorinated oils. Fatty alcohol, fatty acids, waxes and gums, notably silicone gums and elastomers can also be used as fats. Non-limiting examples of suitable emulsifiers and co-emulsifiers include polyglycerol fatty acid esters, sucrose fatty acid esters, sorbitane fatty acid esters, oxyethylene sorbitan fatty acid esters, PEG fatty alcohol ethers, glycerol fatty acid esters, alkyl sulphates, alkyl ether sulphates, alkyl phosphates, alkyl polyglucosides and dimethicone copolyols.
Non-limiting examples of suitable hydrophilic gelling include carboxyvinyl polymers (carbomer), acrylic copolymers such as acrylate/alkylacrylate copolymers, polyacrylamids, polysaccharides such as xanthan gum, guar gum, natural gums such as cellulose gum and derivatives, clays and 2-acrylamido-2-methylpropane acid copolymers.
Non-limiting examples of suitable lipophilic gelling agents include modified clays such as bentones, fatty acid metal salts, hydrophobic silica and ethylcellulose.
Non-limiting examples of suitable fillers include talc, kaolin, mica, serecite, magnesium carbonate, aluminum silicate and organic powders such as nylon.
Non-limiting examples of suitable dyestuffs include lipophilic dyes, hydrophilic dyes, pigments and mother-of-pearl commonly used in cosmetic or dermatological compositions, and their mixtures.
Non-limiting examples of suitable neutralizers include soda, triethanolamine, aminomethyl propanol and potassium hydroxide.
Non-limiting examples of suitable penetration enhancing agents include alcohols and glycols (ethanol and propylene glycol), ethoxydiglycol, alcohols and fatty acids (oleic acid), fatty acid esters and dimethyl isosorbide.
Non-limiting examples of preservatives compatible with cosmetic and pharmaceutical compositions include benzoic acid, its salts and esters, sorbic acid and its salts, parabens and their salts, triclosan, imidazolidinyl urea, phenoxyethanol, DMDM hydantoin, diazolidinyl urea and chlorphenesin.
Conventionally, the filters are UVA and UVB filters. Non-limiting examples of suitable UVA and UVB filters include organic filters such as benzophenone-3, butyl methoxydibenzoyl methane, octocrylene, octyl methoxycinnamate, 4-methylbenzylidene camphor, octyl salicylate, terephthalylidene dicamphor sulfonic acid and drometrizole trisiloxane, and non-organic filters such as titanium oxide and zinc oxide.
Non-limiting examples of suitable solvents include water, ethanol, glycerin, propylene glycol, butylene glycol and sorbitol. The quantities of these various additives are those conventionally used in pharmaceutical preparations as is known to a person skilled in the art.
Methods and uses
According to an aspect of the present invention, there is provided herein a method for treating acetaminophen intoxication in a subject in need thereof, the method comprising administering the combinations of protective agents disclosed herein. In some embodiments, isolated isosilybin B, alone or in combination with one or more one more protective agents selected from the group consisting of MSM, phosphatidylcholine^ Cys) Se-Cys, RibCys, NAC, AD4, Met and SAM are administered. In some embodiments, two or more protective agents are administered, selected from the group consisting of NAC, AD4, Cys, Se-Cys, Met, RibCys phosphatidylcholine, MSM, and isosilybin B.
The administered protective agents may be present in the same of different compositions. The administered protective agents may be administered concurrently or sequentially. The administered protective agents may be administered via the same or different routes of administration.
The compositions and methods of the present invention are typically employed for the treatment of a mammal, preferably a human.
The acetaminophen intoxication includes both acute and chronic intoxication.
According to some conventions, acute intoxication may be defined as consuming total > 150 mg/kg (about 7.5 g in adults) within 24 h. This equals 24 regular-strength tablets over 8 hours or less. However any other dose, including even one pill, may induce toxicity.
Chronic intoxication may occur as a result of long period of low dose consumption, excessive use or repeated overdose.
The symptoms of intoxication may include abdominal pain, appetite loss, coma, convulsions, diarrhea, irritability, jaundice, nausea, sweating, pallor, upset stomach and/or vomiting. Symptoms may also include elevations in the plasma levels of hepatic enzymes and bilirubin, prolongation of the prothrombin time, hypoglycemia and/or lactic acidosis.
Information about acute and chronic acetaminophen intoxication can be found, for example, in The Merck Manual (available at www.merckmanuals.com).
In some embodiments, administration of the compositions as disclosed herein provides a prolonged therapeutic window compared with the conventional, commercially available treatment, meaning that they are effective for a longer period of time following overdose or intoxication. In some embodiments, the therapeutic efficacy is maintained up to 10 hours or more, up to 10-15 hours, up to 15-24 hours, up to 24 hours or more, up to 48 hours or more.
The present invention further provides the use of the protective agents described herein, for the manufacture of a medicament for the treatment of drug intoxication. In some embodiments, the drug is acetaminophen.
The following examples are presented in order to more fully illustrate certain embodiments of the invention. They should in no way, however, be construed as limiting the broad scope of the invention. One skilled in the art can readily devise many variations and modifications of the principles disclosed herein without departing from the scope of the invention.
EXAMPLES
Mice: Male C57BL/6, 9-10 or 11-12 weeks old
Reagents:
Phosphatidylcholine (PC) - PhosChol®, Nutrasal (Nutrasal.Com, Westbrook, ME 04092, made in USA), liquid, kept in room temperature (R/T). Concentration: 3000mg PC per 5ml= 600mg/ml. The solution was diluted to 60mg/ml (stock solution).
N-acetyl-para-aminophenol (APAP)- Tiptipot®, CTS group (made in Israel), liquid.
N-acetylcysteine (NAC) - Flumil™ 2gr, antidote 20% (Pharmazam), 10 ml.
Isosilybin B (IB) - Sigma, Israel, Cat. No. 59527. 5 mg was emulsified in ΙΟΟΟμΙ of DDW (di-distilized water) to yield a 5mg/ml solution which was kept in 4°C.
Methylsulfonylmethane (MSM) - 99.9% pure LIGNISUL MSM -CRYSTAL HEALTH, powder (made in USA) was dissolved in phosphate buffer saline (PBS).
L- Cysteine (Cys): Sigma, Israel (powder, 25gr) dissolved with DDW to 25 mg/ml, PH=5.16.
L-Methionine (Met): Sigma, Israel (powder, 25gr) dissolved with DDW to 50mg/ml. PH=5.84 Example 1 - Isosilvbin B (IB)
IB was tested for its effect on toxicity induced by APAP in comparison to NAC using the following procedure: Mice were allocated into four groups of six mice each, as described in Table 1 below. The mice received sub-lethal hepatotoxic doses of APAP, causing liver damage equivalent to consumption of approximately 10 gr APAP (20 caplets of 500 mg) per 70kg man.
Table 1- Groups and treatment regimens
Figure imgf000025_0001
Mice were treated according to the following procedure:
Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP, APAP+IB or APAP+NAC 15h hours later (9:00 next morning, Day 1) by gavage administration. A total of 350μ1 were administered, containing the different components in the desired concentrations in PBS. Food was returned 2h after APAP administration (Day 1). For all groups, mice were weighted daily, and blood samples were taken 24h hours after APAP administration (Day 2). Serum levels of the liver enzymes aspartate aminotransferase (AST) and alanine aminotransferase (ALT) ALT were measured using a Reflovet® Plus clinical chemistry analyzer (Roche Diagnostics, GmbH, Mannheim, Germany). Serum samples were diluted before the measurements, as needed.
Table 2 presents the effect of each compound on the serum levels of the liver enzymes ALT and AST.
Table 2 -Serum levels of liver enzymes (IU/L)
ALT AST
Group
(average + std.dev.) (average + std.dev.)
18,920.00 +
4 mg APAP 14,360 ± 4,370.52
12,192.04
4 mg APAP+
513.63± 644.37 781.33 + 402.23
3 mg NAC
Figure imgf000026_0001
As can be seen from the above table, IB showed a protective effect when administered with APAP, leading to lower ALT/AST serum levels compared to administration of APAP alone (about 40% and 75% reduction, respectively, compared to administration of APAP only (p<0.05). Advantageously, this effect was observed for relatively low doses of IB, ranging from 0.1-0.4 mg/mouse.
In addition, mice that received APAP with IB showed lower body weight loss between Day 0 and Day 2, compared to mice that received APAP only (p<0.01).
Example 2 - Comparative
The following combinations were tested using the same procedure described in Example 1 above, and compared to 4 mg APAP+ 3 or 3.5 mg NAC:
4 mg APAP+ 0.1 mg Isosilybin A (IA)
4 mg APAP+ 0.2 mg IA
4 mg APAP+ 0.4 mg IA
4 mg APAP+ 0.1 mg silybin B
4 mg APAP+ 0.4 mg silybin B
The results are summarized in Tables 3 and 4.
Table 3 -Serum levels of liver enzymes (IU/L)
ALT AST
Group
(average_+ std.dev.) (average_+ std.dev.)
4 mg APAP 5,617.50 + 1,832.77 3,672.67 + 1,965.99
4 mg APAP+ 3 mg
1,050.25 + 1,248.13 2,717.33 + 3,314.47
NAC
4 mg APAP+ 0.4 mg
7,010.00 + 1,952.85 4,720.67 + 1,909.82
IA
4 mg APAP+ 0.2 mg
6,976.75 + 4,473.26 5,349.33 + 3,537.58
IA
4 mg APAP+ 0.1 mg
8,002.50 + 2,122.65 5,468.00 + 1,986.15
IA Table 4 -Serum levels of liver enzvmes(IU/L)
Figure imgf000027_0001
As can be seen from the above tables, administration of additional ingredients from the milk thistle extract, namely, Isosilybin A and silybin B was shown to be ineffective in reducing liver enzymes serum levels.
Example 3 - IB in combination with NAC
A combination of IB and NAC was tested for its effect on toxicity induced by APAP using the same procedure described in Example 1 above. Mice were allocated into four groups of six mice each, as detailed in Table 5 below. Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP, APAP+NAC or APAP+NAC+IB 15 hours later (9:00 next morning, Day 1) by gavage administration. Food was returned 2h after APAP administration (Day 1).
Table 5 - Groups and treatment regimens
Figure imgf000027_0002
For all groups, mice were weighted daily, and blood samples were taken 24h hours after APAP administration (Day 2) for liver enzyme tests. In addition, liver biopsies were performed. For histopathology, livers from individual mice were fixed in 10% formaldehyde solution and kept at room temperature until use. The tissue blocks were then embedded in paraffin, 5-mm sections were cut and stained with hematoxylin and eosin (H&E) for morphological examination. Figure 1 shows representative histological liver sections from mice taken 24h hours after APAP administration (X40 magnification). In the APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
Table 6 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST.
Table 6 -Serum levels of liver enzymes (IU/L)
Figure imgf000028_0001
As can be seen from the above table, administration of APAP with a combination of 3.5 mg NAC+IB, either 0.1 or 0.4 mg, resulted in over 97% and 91% reduction in the serum levels of ALT and AST, respectively, compared to the administration of APAP only (p< 0.0001). The combinations containing IB were significantly more effective than NAC alone (p< 0.05).
In addition, mice that received APAP with NAC+IB showed a significantly higher increase in body weight between Day 1 and Day 2, compared to mice that received APAP only (p<0.05).
Example 4- Phosphatidylcholine (PC) in combination with NAC
Mice were allocated into four groups of six mice each, as described in Table 7 below. Mice were treated according to the following procedure:
Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP, APAP+NAC or APAP+NAC+PC 15h hours later (9:00 next morning, Day 1) by gavage administration as follows: Group A: APAP
Group B: NAC + APAP
Group C: NAC +APAP , then 1.5 hours later: PC
Group D: NAC +APAP, then 3 hours later: PC
For all groups, food was returned 3.5h after APAP administration.
Table 7 - Groups and treatment regimens
Figure imgf000029_0002
PC was administered 1.5 hours after APAP+NAC administration
*PC was administered 3 hours after APAP+NAC administration
For all groups, mice were weighted daily, and blood samples were taken 24h hours after APAP administration (Day 2) for liver enzyme tests. In addition, liver biopsies were performed.
Figure 2 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification). In APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
Table 8 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 9 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
Table 8-Serum levels of liver enzymes (IU/L)
Figure imgf000029_0001
3 mg NAC+
0.7mg PC after 1.5 h
4 mg APAP+
3 mg NAC+ 193.50 + 131.12 439.33 ± 195.63 0.7mg PC after 3 h
As can be seen from the above table, administration of APAP with a combination of NAC+0.7 mg PC after 1.5 or 3 hrs resulted in about 96-97% and 86-88% reduction in the serum levels of ALT and AST, respectively, compared to the administration of APAP only (p< 0.01).
Table 9 - Effect on body weight
Figure imgf000030_0001
As can be seen from the above table, mice that received APAP only showed a 6% decrease in their body weight during the two days of the experiment. The combination of NAC+PC (administered after 1.5 or 3 hours) almost completely prevented body weight loss, and was significantly more effective than NAC alone (-99% versus -96%, p<0.00001).
Additional measurements have shown that administration of 0.8 mg/mouse PC 2-4 hours following APAP+NAC administration is also effective in reducing serum levels of ALT and AST and preventing body weight loss.
In another experiment testing NAC and PC, the following groups were evaluated:
Table 10- Groups and treatment regimens
Group N Treatment Dosage Admin. fasting
(for treatment)
A 6 APAP 4 mg/mouse PO yes
B 6 APAP+ APAP - 4 mg/mouse PO yes
NAC NAC - 3.5 mg/mouse c 6 APAP+ APAP - 4 mg/mouse PO yes
PC+NAC NAC - 3.5 mg/mouse
PC - 0.9 mg/mouse*
PC was administered 3 hours after APAP+NAC administration
Figure 3 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification). In APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
Table 11 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 12 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2 and by the difference in the mice body weight between Day 1 (immediately after administration of APAP with the different combinations and before food was returned) and Day 2.
Table 11 -Serum levels of liver enzymes (IU/L)
Figure imgf000031_0002
As can be seen from the above table, administration of APAP with a combination of NAC+0.9 mg PC after 3 hrs resulted in about 97% and 89% reduction in the serum levels of ALT and AST, respectively, compared to the administration of APAP only (p< 0.0001). The administration of NAC alone resulted in about 97% and 85% reduction in the serum levels of ALT and AST, respectively, compared to APAP only (p< 0.0001).
Table 12 - Effect on body weight
Figure imgf000031_0001
4 mg APAP+ 2.38
109%
3.5 mg NAC+
0.9 mg PC after 3 h
As can be seen from the above table, mice that received APAP with NAC+PC 0.9 mg after 3 hours showed a significantly higher increase in body weight between Day 1 and Day 2, compared to mice that received APAP only (p<0.01). Administration of NAC alone did not show a significant effect.
In a further experiment testing NAC and PC, the following groups were evaluated:
Table 13- Groups and treatment regimens
Figure imgf000032_0001
PC was administered 3 hours after APAP+NAC administration
Table 14 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 15 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2 and by the difference in the mice body weight between Day 1 (immediately after administration of APAP with the different combinations and before food was returned) and Day 2.
Table 14 -Serum levels of liver enzymes (IU/L)
ALT AST
Group
(average ± std.dev.) (average + std.dev.)
4 mg APAP 5,739.25 ± 2,536.44 4,374.00 + 2,384.21
4.6 mg APAP 7,772.50 + 1 ,771.00 6,310.00 + 2,127.07
4 mg APAP+
3.5 mg NAC 937.00 + 1,280.82 1,963.60 + 2,563.99
0.9 mg PC after 3 h
4.6 mg APAP+
3.5 mg NAC 230.00 + 38.80 485.53 + 168.47 0.9 mg PC after 3 h Table 15 - Effect on body weight
Figure imgf000033_0001
As can be seen from the above tables, administration of APAP, either 4 or 4.6 mg, with a combination of f NAC+0.9 mg PC after 3 hrs resulted in a remarkable reduction in the serum levels of ALT and AST, and a significant higher increase in body weight between Day 1 and Day 2 compared to the administration of APAP only (p< 0.01)
Example 5 - Methylsulfonylmethane (MSM) in combination with NAC
MSM in combination with NAC was tested for its effect on toxicity induced by APAP using the same procedure described in Example 1 above. Mice were allocated into three groups of six mice each, as detailed in Table 16 below. Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP, APAP+NAC or APAP+NAC+MSM 15 hours later (9:00 next morning, Day 1) by gavage administration. Food was returned 3.5h after APAP administration (Day 1).
Table 16- Groups and treatment regimens
Figure imgf000033_0002
Table 17 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 18 presents the effect of each combination on body weight, as determined by the difference in the mice body weight between Day 1 (immediately after administration of APAP with the different combinations and before food was returned) and Day 2.
Figure 4 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification). In APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
Table 17 -Serum levels of liver enzymes (IU/L)
Figure imgf000034_0001
As can be seen from the above table, administration of APAP with a combination of 3.5 mg NAC+1.5 mg MSM resulted in about 98% and 91% reduction in the serum levels of ALT and AST, respectively, compared to the administration of APAP only (p< 0.001). The combination of NAC+MSM appeared to be more effective than NAC alone.
Table 18 - Effect on body weight
Figure imgf000034_0002
As can be seen from the above table, mice that received APAP with 3.5 mg NAC+MSM 1.5 mg showed a significantly higher increase in body weight between Day 1 and Day 2, compared to mice that received APAP only (p<0.01). The combination of NAC+MSM appeared to be more effective than NAC alone. Example 6 - Combination of NAC. MSM. PC and IB
This combination was tested for its effect on toxicity induced by APAP using the procedure described in Example 1 above with several modifications, as described below. Mice were allocated into four groups of six mice each, as detailed in Table 19 below. Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered APAP 15 hours later (9:00 next morning, Day 1) by gavage administration. NAC was administered PO with APAP. MSM+IB were administered 1.5h later. Following another 1.5h, PC was administered. Food was returned 3.5h after APAP administration (Day 1). Weight was determined three times: on Day 0 before food was taken, after over-night fasting (Day 1, just before APAP administration) and before sacrifice (Day 2, 24 hours after APAP administration).
Table 19 - Groups and treatment regimens
Figure imgf000035_0002
Table 20 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 21 presents the effect of each combination on body weight, as determined by the difference/ratio in the mice body weight between Day 1 and 2.
Table 20 -Serum levels of liver enzymes (IU/L)
Figure imgf000035_0001
Figure imgf000036_0001
Table 21 - Effect on body weight
Figure imgf000036_0002
As can be seen from the above tables, administration of 7 mg APAP per mouse resulted in severe intoxication. Administration of NAC in combination with IB, MSM and PC resulted in a major reduction of the serum levels of ALT and AST, and a significantly higher increase in body weight between Day 1 and Day 2 compared to the administration of APAP only. This combination was more effective than NAC alone.
Example 7 - Combination of MSM and IB
This combination was tested for its effect on toxicity induced by APAP using the procedure described in Example 1 above.
Table 22 - Groups and treatment regimens
Group N Treatment Dosage Admin. fasting
(for treatment)
A 6 APAP 4 mg/mouse PO yes
B 6 APAP+ MSM APAP - 4 mg/mouse PO yes
MSM - 0.15
mg/mouse
C 6 APAP+ IB APAP - 4 mg/mouse PO yes
0.1 mg/mouse IB D 6 APAP+ APAP - 4 mg/mouse PO yes
MSM+IB MSM - 0.15
mg/mouse
0.1 mg/mouse IB
Table 23 -Serum levels of liver enzymes (IU/L)
Figure imgf000037_0001
As can be seen from the above table, a combination of MSM and IB showed significant reduction of the serum levels of ALT and AST (p<0.05). The combination was superior to each of the compounds alone, which did not show a significant effect.
In another experiment testing MSM and IB, the following groups were evaluated:
Table 24 - Groups and treatment regimens
Figure imgf000037_0002
Table 25 -Serum levels of liver enzymes (IU/L)
ALT AST
Group
(average + std.dev.) (average + std.dev.)
4mg APAP 6,896.00_+ 2,998.94 4,303.33 + 2,163.30
4mg APAP+
8,502.00 + 2,186.33 5,076.00 + 1,616.62 0.25 mg MSM
Figure imgf000038_0001
Once again, the combination of MSM and IB in group D showed significant reduction of the serum levels of ALT and AST (p<0.05). The combination was much superior to each of the compounds alone, which did not show any meaningful effect.
In order to determine that the combination of MSM+IB does not interfere with the anti-inflammatory effect of APAP, splenocytes were isolated from mice from each group and tested for their IFN-γ secretion. After mice were sacrificed, spleens from five mice from each group were taken and splenocytes were isolated by the following procedure:
Isolation of splenocytes: Spleens were stored in RPMI-1640 medium supplemented with FCS. Spleens were crushed through a 70-μιη nylon cell strainer (Falcon) and centrifuged (1,250 rpm for 7 min.). Red blood cells were lysed in 1 ml of cold 155 mM ammonium chloride lysis buffer. Splenocytes were washed and resuspended in 1 ml of RPMI supplemented with FCS. The viability of cells as assessed by trypan blue exclusion exceeded 90%.
Next, splenocytes (lxlO6 per mouse, per well, in duplicates) were moved into a 24 well plate containing RPMI-1640 supplemented with 10% FCS, 1% penicillin/streptomycin (P/S), 1 % glutamine, 1 % sodium pyruvate and 1% non-essential amino acid. All splenocytes were activated with 5μg of Concanavalin A (purchased from MP Biomedicals, Ohio, USA), for 20 h at 37°C. After incubation cells from all wells were moved to Eppendorff tubes and centrifuged for 5 minutes. Then supernatants were collected and freezed in -80°C until assayed for IFN-γ by ELISA.
Cytokine determination: Levels of IFN-γ were determined by "sandwich" ELISA using commercial kit according to the manufacturer's instructions (Quantikine, R&D Systems, Minneapolis, MN, USA).
Figure 5 shows IFN-γ levels secreted by ConA-activated splenovytes derived from each group of mice. As can be seen in the figure, the combination of MSM+IB had no effect on the secretion of IFN-γ from in vitro ConA-activated splenocytes (259, 255, 229, 253 pg/ml of IFN-γ, for groups A, B, C and D, respectively, p=NS). Thus, the results have shown that oral administration of the above combination of MSM+IB with APAP is associated with a similar anti-inflammatory effect to that of APAP, while significantly alleviating the APAP-induced liver injury.
In another experiment the effect of the IB+MSM combination on APAP absorption was tested. Mice treated with lOmg of APAP alone or in combination with 0.1 mg IB + 0.25 mg MSM were followed for APAP serum levels. The results have shown that the combination of IB+MSM had no effect on the bioavailability of APAP. Serum levels of APAP in mice treated with 10 mg of APAP alone were similar to mice treated with APAP+IB+MSM (129 vs. 149 ug/ml, respectively. p=NS).
Example 8 - Combinations with Cvs and Met
Mice were allocated into four groups of six mice each, as described in Table 26 below. The mice received sub-lethal hepatotoxic doses of APAP (4.6mg/mouse). The effect of the different combinations of compounds on toxicity induced by APAP was tested using the procedure described in Example 1 above.
Table 26- Groups and treatment regimens
Figure imgf000039_0001
Figure 6 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification). In APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups. Table 27 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 30 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
Table 27 -Serum levels of liver enzymes (IU/L)
Figure imgf000040_0002
As can be seen from the above table, administration of APAP in combination of NAC resulted in about 95-97% reduction in the serum levels of ALT and AST, compared to the administration of APAP only (p< 0.005). Similarly, administration of APAP and NAC substitutes (Cys or Met) in combination with PC, MSM and IB resulted in a comparable decrease in liver enzymes.
Table 28 - Effect on body weight
Figure imgf000040_0001
As can be seen from the above table, mice that received APAP only showed more than 8% decrease in their body weight during the two days of the experiment. The combination of NAC substitutes with PC, MSM and IB almost completely prevented body weight loss, and was more effective as NAC alone ( p<0.01).
In another experiment the following groups were evaluated:
Table 29- Groups and treatment regimens
Figure imgf000041_0001
*MSM and IB were administered 1.5 hours after APAP+NAC or after APAP+ Cys or after APAP+ Met, were administered.
**PC was administered 3 hours after APAP+NAC or after APAP+Cys or after APAP+ Met; were administered.
Mice were treated according to the following procedure:
Food was taken on the afternoon (18:00) of Day 0. Mice were orally administered various preparations (as mentioned in Table 31) 15h hours later (9:00 next morning, Day 1) by gavage administration as follows:
Group A: APAP
Group B: NAC + APAP. After 1.5h, MSM + IB. After another 1.5h, PC
Group C: APAP + Cys. After 1.5h, MSM + IB. After another 1.5h, PC
Group D: APAP + Met. After 1.5h, MSM + IB). After another 1.5h, PC
Figure 7 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification). In APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
Table 30 presents the effect of each combination on the serum levels of the liver enzymes ALT and AST. Table 33 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
Table 30 -Serum levels of liver enzymes (IU/L)
Figure imgf000042_0002
As can be seen from the above table, administration of APAP with a combination of NAC with later on administration of MSM, IB and afterwards PC, resulted in about 95- 96% reduction in the serum levels of ALT and AST, compared to the administration of APAP only (p< 0.07). When NAC was replaced by either Cys or Met, the effect on liver enzymes even increased to 98-99% reduction in ALT levels and 97-98% reduction in AST levels compared to the administration of APAP only (p<0.005).
Table 31- Effect on body weight
Figure imgf000042_0001
As can be seen from the above table, mice that received APAP only showed more than 8% decrease in their body weight during the two days of the experiment. The combination of NAC with PC, MSM and IB lowered body weight loss to 96.13%. When NAC was replaced by either Cys or Met, the effect on body weight was similar and even slightly increased to 97-98% (p<0.007).
In another experiment the following groups were evaluated:
Table 32- Groups and treatment regimens
Figure imgf000043_0001
Mice were treated according to the procedure describe in Example 1 above.
Figure 8 shows representative histological liver sections from mice taken 24h hours after APAP administration n (X40 magnification). In APAP group, diffuse damage to hepatocytes can be easily seen (hemorrhagic congestion and extensive centrilobular necrosis). Improved liver architecture with no evidence for cellular damage and necrosis can be seen in all treated groups.
Table 33 presents the effect of various treatments on the serum levels of the liver enzymes ALT and AST. Table 36 presents the effect of each combination on body weight loss, as determined by the ratio of the mice body weight between Day 0 and Day 2.
Table 33 -Serum levels of liver enzymes (IU/L)
ALT AST
Group
(average_± std.dev.) (average_+ std.dev.)
APAP
9002.50 + 4211.03 5684.00+ 2307.64
APAP+ NAC
1175.26 + 1552.79 821.93 + 766.57 APAP+ Cys
196.06 + 112.80 369.47 + 117.51
APAP + Met
147.84 ± 49.24 332.97+ 100.52
As can be seen from the above table, administration of APAP with NAC resulted in about 85-87% reduction in the serum levels of ALT and AST compared to administration of APAP alone. Administration of APAP with Cys or Met resulted in a greater reduction in the liver enzymes, up to about 94-98% reduction compared to APAP alone (p<0.0005).
Table 34- Effect on body weight
Figure imgf000044_0001
As can be seen from the above table, mice that received APAP only showed more than 6% decrease in their body weight during the two days of the experiment. The combination of APAP with NAC resulted in lower weight loss, to 98.08%. When NAC was replaced by either Cys or Met, the effect on body weight was similar (97-99% p<0.007).
The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying current knowledge, readily modify and/or adapt for various applications such specific embodiments without undue experimentation and without departing from the generic concept, and therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not of limitation. The means, materials, and steps for carrying out various disclosed chemical structures and functions may take a variety of alternative forms without departing from the invention.

Claims

1. A pharmaceutical composition comprising isolated isosilybin B, and optionally further comprising at least one additional protective agent selected from the group consisting of methylsulfonylmethane (MSM), phosphatidylcholine 5 cysteine (Cys), seleno- cysteine (Se-Cys), ribose-cysteine (RibCys), N-acetylcysteine (NAC), N- acetylcysteine-amide (AD4), methionine (Met) and S-adenosylmethionine (SAM), wherein said pharmaceutical composition is effective in treating or preventing drug- induced toxicity.
2. The pharmaceutical composition of claim 1, comprising isolated isosilybin B and further comprising (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
3. The pharmaceutical composition of claim 1, wherein the drug is acetaminophen.
4. The pharmaceutical composition of claim 1, further comprising a drug with potential toxicity.
5. The pharmaceutical composition of claim 4, wherein the drug is acetaminophen.
6. The pharmaceutical composition of claim 5, further comprising at least one additional drug selected from the group consisting of an opioid analgesic, a decongestant, a cough suppressant, an antihistamine and an expectorant.
7. The pharmaceutical composition of claim 1, formulated for a route of administration selected from the group consisting of oral, rectal, intravenous intramuscular, intradermal, subcutaneous and intranasal.
8. A method for treating drug intoxication in a subject in need thereof, the method comprising administering to the subject isolated isosilybin B, and optionally at least one additional protective agent selected from the group consisting of MSM, phosphatidylcholine 5 Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM.
9. The method of claim 8, comprising administering to the subject isolated isosilybin B and further administering (i) one or more protective agents selected from the group consisting of Cys, Se-Cys, RibCys, NAC, AD4, Met and SAM; and (ii) one or more protective agents selected from the group consisting of MSM and phosphatidylcholine.
10. The method of claim 8, wherein the isolated isosilybin B and the at least one additional protective agent are within a single pharmaceutical composition.
11. The method of claim 8, wherein the isolated isosilybin B and the at least one additional protective agent t are within separate pharmaceutical compositions.
12. The method of claim 8, wherein the isolated isosilybin B and the at least one additional protective agent are administered concurrently.
13. The method of claim 8, wherein the isolated isosilybin B and the at least one additional protective agent are administered sequentially.
14. The method of claim 8, wherein the isolated isosilybin B and the at least one additional protective agent are administered via the same route of administration.
15. The method of claim 8, wherein the isolated isosilybin B and the at least one additional protective agent are administered via different routes of administration.
16. The method claim 8, wherein administration is performed via a route of administration selected from the group consisting of oral, rectal, intravenous, intramuscular, intradermal, subcutaneous and intranasal.
17. The method claim 7, wherein the drug is acetaminophen.
18. The method of claim 17, wherein the drug is acetaminophen in combination with at least one additional drug selected from the group consisting of an opioid analgesic, a decongestant, a cough suppressant, an antihistamine and an expectorant.
19. The method of claim 17, wherein the acetaminophen intoxication is acute intoxication.
20. The method of claim 17, wherein the acetaminophen intoxication is chronic intoxication.
21. A composition comprising isolated isosilybin B, and optionally at least one more protective agent selected from the group consisting of MSM, phosphatidylcholine^ Cys) Se-Cys, RibCys, NAC, AD4, Met and SAM, for use in the treatment of drug intoxication.
22. The composition of claim 21, wherein the drug is acetaminophen.
23. The composition of claim 21, formulated for administration via a route of administration selected from the group consisting of oral, rectal, intravenous, intramuscular, intradermal, subcutaneous and intranasal.
PCT/IL2013/050446 2012-05-24 2013-05-23 Compositions comprising isosilybin b for amelioration and prevention of drug- induced toxicity WO2013175479A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP13793975.7A EP2854798A4 (en) 2012-05-24 2013-05-23 Compositions comprising isosilybin b for amelioration and prevention of drug- induced toxicity
US14/551,724 US9474763B2 (en) 2012-05-24 2014-11-24 Compositions and methods for amelioration and prevention of drug-induced toxicity

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261651005P 2012-05-24 2012-05-24
US61/651,005 2012-05-24
US201261654971P 2012-06-04 2012-06-04
US61/654,971 2012-06-04

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/551,724 Continuation US9474763B2 (en) 2012-05-24 2014-11-24 Compositions and methods for amelioration and prevention of drug-induced toxicity

Publications (1)

Publication Number Publication Date
WO2013175479A1 true WO2013175479A1 (en) 2013-11-28

Family

ID=49623253

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2013/050446 WO2013175479A1 (en) 2012-05-24 2013-05-23 Compositions comprising isosilybin b for amelioration and prevention of drug- induced toxicity

Country Status (3)

Country Link
US (1) US9474763B2 (en)
EP (1) EP2854798A4 (en)
WO (1) WO2013175479A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3302456A4 (en) * 2015-06-05 2018-12-19 Glenn A. Goldstein Use of n-acetylcysteine amide in the treatment of acetaminophen overdose

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022269026A1 (en) 2021-06-25 2022-12-29 Forschungsgesellschaft Für Arbeitsphysiologie Und Arbeitsschutz E. V. Treating acetaminophen overdose

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4868114A (en) 1987-08-05 1989-09-19 Regents Of The University Of Minnesota Method for elevating glutathione levels
US6555141B1 (en) 1998-02-27 2003-04-29 Nutramax Laboratories, Inc. L-ergothioneine, Milk thistle, and S-adenosylmethionine for the prevention, treatment and repair of liver damage
US6566401B2 (en) 2001-03-30 2003-05-20 The Board Of Trustees Of The Leland Stanford Junior University N-acetylcysteine compositions and methods for the treatment and prevention of drug toxicity
US20070021376A1 (en) 2005-07-21 2007-01-25 Suracell, Inc. Supplement composition and method of use in enhancement of methylation process
US7238373B2 (en) 2003-04-04 2007-07-03 Nutritox Llc Nutritional supplement
WO2009097874A1 (en) 2008-02-07 2009-08-13 Velleja Research S.R.L. Amino acid formulations comprising cysteine, methionine and/or serine for the prevention of paracetamol-induced liver damage
WO2011044230A2 (en) 2009-10-06 2011-04-14 Goldstein Glenn A N-acetylcysteine amide (nac amide) for the treatment of diseases and conditions
US20110124718A1 (en) 2009-11-24 2011-05-26 Mccain Mark D Reduction Of Acetaminophen Toxicity By Dietary Milk Thistle Extract
US20120022161A1 (en) 2010-07-21 2012-01-26 Cumberland Pharmaceuticals, Inc. Acetycysteine compositions and methods of use thereof
US8148356B2 (en) 2005-08-24 2012-04-03 Cumberland Pharmaceuticals, Inc. Acetylcysteine composition and uses therefor

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US6576267B2 (en) 2000-02-23 2003-06-10 Bioselect Innovations, Inc. Composition and method for treating the effects of diseases and maladies
US6495170B1 (en) * 2000-08-16 2002-12-17 N. V. Nutricia Method of increasing the presence of glutathione in cells
CN1956726B (en) * 2004-03-23 2011-04-13 生命线营养健康公司 Composition and method for releasing mammal inflammation and oxidation stress

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4868114A (en) 1987-08-05 1989-09-19 Regents Of The University Of Minnesota Method for elevating glutathione levels
US6555141B1 (en) 1998-02-27 2003-04-29 Nutramax Laboratories, Inc. L-ergothioneine, Milk thistle, and S-adenosylmethionine for the prevention, treatment and repair of liver damage
US6566401B2 (en) 2001-03-30 2003-05-20 The Board Of Trustees Of The Leland Stanford Junior University N-acetylcysteine compositions and methods for the treatment and prevention of drug toxicity
US7238373B2 (en) 2003-04-04 2007-07-03 Nutritox Llc Nutritional supplement
US20070021376A1 (en) 2005-07-21 2007-01-25 Suracell, Inc. Supplement composition and method of use in enhancement of methylation process
US8148356B2 (en) 2005-08-24 2012-04-03 Cumberland Pharmaceuticals, Inc. Acetylcysteine composition and uses therefor
WO2009097874A1 (en) 2008-02-07 2009-08-13 Velleja Research S.R.L. Amino acid formulations comprising cysteine, methionine and/or serine for the prevention of paracetamol-induced liver damage
WO2011044230A2 (en) 2009-10-06 2011-04-14 Goldstein Glenn A N-acetylcysteine amide (nac amide) for the treatment of diseases and conditions
US20110124718A1 (en) 2009-11-24 2011-05-26 Mccain Mark D Reduction Of Acetaminophen Toxicity By Dietary Milk Thistle Extract
US20120022161A1 (en) 2010-07-21 2012-01-26 Cumberland Pharmaceuticals, Inc. Acetycysteine compositions and methods of use thereof

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"Drug Facts and Comparisons", 1993, FACTS AND COMPARISONS, INC.
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
ANTHONY S. TRAVIS: "Zvi Rappoport. The chemistry of Anilines Part 1", 2007, WILEY, article "Manufacture and uses of the anilines: A vast array of processes and products", pages: 764
CHASSAING ET AL., J CHROMATOGR B BIOMED SCI APPL, vol. 735, no. 2, 1999, pages 219 - 27
DVORAK ET AL., TOXICOLOGY LETTERS, vol. 3, 2003, pages 201 - 212
ELMAR FRIDERICHS; THOMAS CHRISTOPH; HELMUT BUSCHMANN: "Ullmann's Encyclopedia of Industrial Chemistry", 2005, WILEY-VCH, article "Analgesics and Antipyretics"
GOODMAN; GILMAN: "The Physician's Desk Reference", 1995, MEDICAL ECONOMICS COMPANY, INC.
POLYAK ET AL., PNAS U.S.A, vol. 107, 2010, pages 5995 - 5999
POLYAK ET AL.: "Identification ofhepatoprotective flavonolignans from Silymarin", PNAS, vol. 107, no. 13, 30 March 2010 (2010-03-30), pages 5995 - 5999, XP055035626 *
REMINGTON; GENNARO, A.: "The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
See also references of EP2854798A4 *
SOUMENDRA DARBAR ET AL.: "Antihepatoprotective potential of Livina, a polyherbal preparation on parcetamol induced hepatotoxicity: a comparison with Silymarin", ASIAN JOURNAL OF PHARMACEUTICAL AND CLINICAL RESEARCH, vol. 4, no. 1, 31 December 2011 (2011-12-31), pages 72 - 77, XP055177096 *
ZDENEK DVORAK ET AL.: "Primary cultures of human hepatocytes as a tool in cytotoxicity studies: cell protection against model toxins by flavonolignans obtained from Silybum marianum", TOXICOLOGY LETTERS, vol. 3, 3 February 2003 (2003-02-03), pages 201 - 212, XP055177099 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3302456A4 (en) * 2015-06-05 2018-12-19 Glenn A. Goldstein Use of n-acetylcysteine amide in the treatment of acetaminophen overdose

Also Published As

Publication number Publication date
EP2854798A1 (en) 2015-04-08
US9474763B2 (en) 2016-10-25
EP2854798A4 (en) 2016-05-11
US20150080348A1 (en) 2015-03-19

Similar Documents

Publication Publication Date Title
AU2011330757B2 (en) Continuous administration of L-dopa, dopa decarboxylase inhibitors, catechol-O-methyl transferase inhibitors and compositions for same
CN102625699B (en) Treatment of portal hypertension and restoration of liver function using L-ornithine phenylacetate
EP2453743B1 (en) N-acetyl cysteine compositions and their use in improving the therapeutic efficacy of acetaminophen
US20200138830A1 (en) Enhancing gaba&#39;s ability to modulate immune responses
ES2356986T3 (en) ANTITUMOR AGENTS.
US9474763B2 (en) Compositions and methods for amelioration and prevention of drug-induced toxicity
WO2010004060A1 (en) Use of cilastatin for reducing nephrotoxicity of different compounds
JP5161472B2 (en) Analgesic composition
JP2009242365A (en) Oral pharmaceutical composition
JP2022019937A (en) Composition for preventing or treating chronic or acute virus infection and/or sepsis in humans or animals
EP2076239A1 (en) Combination therapy
US20120071498A1 (en) Therapeutic composition to treat lesions caused by herpes simplex virus
US8927601B2 (en) Uses of N-butylidenephthalide in treating a liver injury and improving liver function
JP6100510B2 (en) Anti-cold medicine
CN102357249A (en) Medicine for inhibiting medicine-resistant tubercle bacillus
KR100379155B1 (en) New analgesic composition
Chávez et al. Are N-Acetylcysteine and Resveratrol Effective Treatments for Liver Disease?
CN106474126B (en) Application of anticancer small molecule compound sorafenib in treating liver echinococcosis
ES2608476B1 (en) COMPOUNDS FOR THE TREATMENT OF DISEASES CAUSED BY PARASITES OF THE LEISHMANIA GENDER
Horoshko Influence of nano-drugs on lipid and protein peroxidation processes in experimental acute renal failure
WO2023215323A1 (en) Intranasal baclofen
AU2021309398A1 (en) A composition for treating helminth infestation in a non-human mammal
JP2021161105A (en) Composition for preventing or treating chronic or acute virus infection and/or sepsis in humans or animals
JP6047004B2 (en) Safe pharmaceutical composition
CN116327746A (en) Application of isoliquiritigenin in preparation of medicine for preventing or treating drug-induced liver injury caused by acetaminophen

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13793975

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013793975

Country of ref document: EP