US20060121132A1 - TNF-alpha production inhibitor comprising kavalactone as an active ingredient - Google Patents

TNF-alpha production inhibitor comprising kavalactone as an active ingredient Download PDF

Info

Publication number
US20060121132A1
US20060121132A1 US11/335,641 US33564106A US2006121132A1 US 20060121132 A1 US20060121132 A1 US 20060121132A1 US 33564106 A US33564106 A US 33564106A US 2006121132 A1 US2006121132 A1 US 2006121132A1
Authority
US
United States
Prior art keywords
tnf
disease
kavalactone
kava
production
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/335,641
Inventor
Yoshinori Asakawa
Sachiko Okabe
Masashi Yamada
Yukie Suma
Hiroto Suzuki
Masayuki Uchida
Natsuko Murata
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Meiji Dairies Corp
Original Assignee
Meiji Dairies Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from JP2000383095A external-priority patent/JP2001316260A/en
Application filed by Meiji Dairies Corp filed Critical Meiji Dairies Corp
Priority to US11/335,641 priority Critical patent/US20060121132A1/en
Publication of US20060121132A1 publication Critical patent/US20060121132A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/67Piperaceae (Pepper family), e.g. Jamaican pepper or kava
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a TNF- ⁇ production inhibitor containing a kavalactone as an active ingredient, and to a preventive, ameliorating, or therapeutic agent for diseases caused by abnormal production of TNF- ⁇ .
  • TNF Tumor Necrosis Factor
  • Kava is a plant found in Fiji and belongs to Piperaceae, Piper L. (nomenclature: Piper Methysticum Forst ., alias: Yangona). Since anesthetic beverages are obtained from the kava root, in Oceania, kava is widely cultivated by privileged people and is used in traditional ceremonies or events ( Chem. Australia. October 377-378 (1987)).
  • kavalactones exert a sedative effect through a mechanism different from those of other sedative drugs which exert sedative effects when being bound to receptors present in the brain ( Planta Med. 65 507-510 (1998)). It has also been reported that kavalactones exert an analgesic effect in a manner different from that of a formulated analgesic drug such as aspirin, and that, unlike morphine, kavalactones are not bound to receptors in the brain (e.g., European Patent Application Laid-Open Nos. 664131 and 523591, and Japanese Kohyo (PCT) Patent Publication No. 5-502457).
  • European Patent Application Laid-Open Nos. 664131 and 523591 and Japanese Kohyo (PCT) Patent Publication No. 5-502457.
  • kava extract exerts an antibacterial effect and is useful for treating Helicobacter pylori infection (German Patent Application Laid-Open No. 19716660), and that the kava extract exerts a neuroprotective effect and is useful for treating brain dysfunction, Alzheimer's disease, brain injury, etc. (e.g., European Patent Application Laid-Open No. 523591, and Japanese Kohyo (PCT) Patent Publication No. 5-502457).
  • the present inventors have performed studies on naturally occurring substances which inhibit production of TNF- ⁇ , and have found that kavalactones contained in kava extract exert an excellent effect of inhibiting TNF- ⁇ production, and that the kavalactones are useful as TNF- ⁇ production inhibitors and as preventive, ameliorating, or therapeutic agents for a variety of diseases caused by abnormal production of TNF- ⁇ .
  • the present invention has been accomplished on the basis of this finding.
  • an object of the present invention is to provide a drug which is endowed with high safety, inhibits TNF- ⁇ production, and is useful as a preventive or therapeutic agent for the aforementioned diseases.
  • the present invention provides a TNF- ⁇ production inhibitor comprising a kavalactone as an active ingredient.
  • the present invention also provides a preventive, ameliorating, or therapeutic agent comprising a kavalactone as an active ingredient for diseases caused by abnormal production of TNF- ⁇ .
  • the present invention further provides a method for the treatment of diseases caused by abnormal production of TNF- ⁇ , which method comprises administering an effective amount of a kavalactone.
  • the present invention further provides use of a kavalactone for the manufacture of a TNF- ⁇ production inhibitor.
  • the present invention provides use of a kavalactone for the manufacture of a medicament for preventing, ameliorating, or treating diseases caused by abnormal production of TNF- ⁇ .
  • the kavalactone is one or more species selected from the group consisting of desmethoxyyangonin, dihydrokavain, kavain, yangonin, methysticin, dihydromethysticin, and 7,8-epoxyyangonin.
  • FIG. 1 shows a graph indicating inhibitory effects of KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 on TNF- ⁇ production in BALB/3T3 cells stimulated by okadaic acid;
  • FIG. 2 shows a graph showing investigation results of inhibitory effects of KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 on TNF- ⁇ production in serum samples, the TNF- ⁇ production having been induced by intraperitoneally administering KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 to BALB/c mice, followed by administration of LPS immediately thereafter.
  • Kavalactone contained, as an active ingredient, in the TNF- ⁇ production inhibitor of the present invention refers to a class of ⁇ -pyrone derivatives contained in the root of kava ( Piper Methysticum G. Forst ) which belongs to Piperaceae, Piper L.
  • Specific examples of the ⁇ -pyrone derivatives include desmethoxyyangonin, dihydrokavain, kavain, yangonin, methysticin, dihydromethysticin, and 7,8-epoxyyangonin, which are represented by the following formulas.
  • ⁇ -pyrone derivatives have a variety of isomers, including geometrical isomers such as cis-isomers and trans-isomers, optical isomers such as d-isomers and l-isomers, and rotational isomers.
  • geometrical isomers such as cis-isomers and trans-isomers
  • optical isomers such as d-isomers and l-isomers
  • rotational isomers any of such isomers can be used, so long as the isomer can exert the effect of inhibiting TNF- ⁇ production.
  • these pyrone derivatives also include racemic modifications and a mixture of diastereomers.
  • examples of particularly preferred kavalactones include desmethoxyyangonin (KAVA-3: compound 1), (+)-dihydrokavain (KAVA-4: compound 2), (+)-kavain (KAVA-5: compound 3), yangonin (KAVA-2: compound 4), (+)-methysticin (KAVA-1: compound 5), (+)-dihydromethysticin (KAVA-6: compound 6), and 7,8-epoxyyangonin (compound 7).
  • ⁇ -pyrone derivatives may be used singly or in combination of two or more species as kavalactone employed in the TNF- ⁇ production inhibitor of the present invention.
  • Kavalactones used in the present invention may be obtained from the kava root by means of a known extraction method, or obtained through synthesis by means of a published method ( Acta Chemica Scandinavica B 30, 7: 613-678, 1976; Planta Med., 64: 504, 1998).
  • kavalactones are obtained through extraction
  • the aforementioned kavalactones separated from kava extract and purified can be used.
  • an extraction fraction containing a plurality of compounds may also be used, so long as the fraction exhibits the effect of inhibiting TNF- ⁇ production.
  • the kavalactones can be used as a TNF- ⁇ production inhibitor for mammals including humans, and as a preventive, ameliorating, or therapeutic agent for a variety of diseases caused by abnormal production of TNF- ⁇ , including cachexia attributed to cancer or infectious diseases, septic shock, chronic rheumatoid arthritis, inflammatory diseases such as ulcerative colitis and Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus (SLE), rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes.
  • cachexia attributed to cancer or infectious diseases including cachexia attributed to cancer or infectious diseases, septic shock, chronic rheumatoid arthritis, inflammatory diseases such as ulcerative colitis and Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lup
  • the amount and frequency of administration vary with pathological conditions, age, weight, manner of administration, and other conditions.
  • the daily dose is typically 0.1-1,000 mg for adults, but the daily dose varies with pathological conditions and other conditions (e.g., 10-500 mg or 30-300 mg).
  • pathological conditions and other conditions e.g., 10-500 mg or 30-300 mg.
  • a daily dose of 200 mg has been reported (Kretschmer “Kavain als Psychopharmkon,” NMW 4/1970, 154-158).
  • the kavalactone according to the present invention is used as a TNF- ⁇ production inhibitor and as a preventive, ameliorating, or therapeutic agent for diseases caused by abnormal production of TNF- ⁇
  • the kavalactone is prepared in the form of a typical pharmaceutical product.
  • the kavalactone is formulated in a form suitable for oral administration or parenteral administration (e.g., intraarticular administration or enteric administration), such as a pharmaceutical composition obtained by mixing the drug of the present invention with a pharmaceutically acceptable carrier (e.g., an excipient, a binder, a disintegrant, a sweetening agent, a flavoring agent, an emulsifying agent, a diluent, or a dissolution promoter) and processing to have a product form of, for example, tablet, pill, powder, granule, capsule, troche, syrup, solution, emulsion, suspension, or injection.
  • a pharmaceutically acceptable carrier e.g., an excipient, a binder, a disintegrant, a sweetening agent, a flavoring agent, an emulsifying agent, a diluent, or a dissolution promoter
  • a pharmaceutically acceptable carrier e.g., an excipient, a binder, a dis
  • excipients include lactose, cornstarch sucrose, glucose, sorbitol, and crystalline cellulose.
  • binders include polyvinyl alcohol, polyvinyl ether, ethyl cellulose, methyl cellulose, gum arabi, tragacanth, gelatin, shellac, hydroxypropyl cellulose, hydroxypropyl starch, and polyvinyl pyrrolidone.
  • disintegrants examples include starch, agar, gelatin powder, crystalline cellulose, calcium carbonate, sodium hydrogencarbonate, calcium citrate, dextran, and pectin.
  • lubricants include magnesium stearate, talc, polyethylene glycol, silica, and hydrogenated vegetable oil. Any pharmaceutically acceptable coloring agent may be used.
  • sweetening and flavoring agents include cocoa powder, menthol, aromatic acids, peppermint oil, borneol, and cinnamon powder. If necessary, tablets or granules may optionally be subjected to sugar coating, gelatin coating, or similar coating.
  • a pH-regulating agent, a buffer, a stabilizer, or a preservative is added to the injections, to thereby prepare agents for subcutaneous injection, intramuscular injection, or intravenous injection, by means of a customary method.
  • Injection preparations may be stored in a container and freeze-dried, to thereby provide solid products, and the solid products may be prepared into injections upon use.
  • a single dose of the injection may be stored in a container, or a plurality of doses may be stored in a single container.
  • Dried kava root (1.0 kg) was subjected to extraction by use of methanol for 16 days.
  • the resultant mixture was subjected to filtration under reduced pressure, and the filtrate was concentrated under reduced pressure, to thereby yield a residue (152.0 g).
  • the entirety of the residue was partitioned by use of water and ethyl acetate (AcOEt), and the resultant ethyl acetate phase was concentrated under reduced pressure, to thereby yield an ethyl acetate extract (76.17 g).
  • the entirety of the extract was added to a silica gel column (product of Merck, 1 kg (70-230 mesh 500 g+230-400 mesh 500 g)) for chromatography by use of n-hexane/ethyl acetate.
  • n-hexane 500 ml
  • 5% ethyl acetate/n-hexane 500 ml
  • 10% ethyl acetate/n-hexane 500 ml
  • 15% ethyl acetate/n-hexane 500 ml
  • 20% ethyl acetate/n-hexane 500 ml
  • 40% ethyl acetate/n-hexane 500 ml
  • 45% ethyl acetate/n-hexane 500 ml
  • BALB/3T3 cells produce TNF- ⁇ .
  • the compounds of the present invention were investigated in terms of inhibitory effect on TNF- ⁇ production.
  • An MEM medium (product of Nissui) containing 10% fetal calf serum (product of Biocell Laboratory) was injected into 12-well multiplates (product of Corning), and BALB/3T3 cells were disseminated at 2 ⁇ 10 5 cells/well.
  • the cells were cultured in a carbon dioxide gas incubator (5% CO 2 , humidified, 37° C.). Subsequently, KAVA-1, KAVA-2, KAVA-3, KAVA-4, or KAVA-5 was added to the wells at a concentration shown in FIG. 1 , and the cells were cultured for one hour. No KAVA compound was added to the control wells.
  • okadaic acid (carcinogenisis promoter isolated from Halichondria okadai ) was added to each well at a final concentration of 0.2 ⁇ M, and culturing was performed for 24 hours. After completion of this culturing, the TNF- ⁇ concentration of the supernatant of each well was measured by means of ELISA system (product of Genzyme). The results are shown in FIG. 1 . In FIG. 1 , the amount of TNF- ⁇ release corresponding to each compound concentration is represented by a percent concentration based on the amount of TNF- ⁇ release measured for the control (100%).
  • KAVA-3 was found to inhibit TNF- ⁇ production to approximately 60% (at 10 ⁇ M) the TNF- ⁇ production of the control; to approximately 22% (at 50 ⁇ M) the TNF- ⁇ production of the control; and to approximately 0% (complete inhibition) (at 100 ⁇ M).
  • KAVA-2 was found to inhibit TNF- ⁇ production to approximately 22% (at 100 ⁇ M).
  • KAVA-5 and KAVA-1 exerted similar inhibitory effects; i.e., exerted inhibition to approximately 39% (at 100 ⁇ M).
  • KAVA-4 exerted no inhibitory effect at concentrations of 50 ⁇ M or less, but exerted inhibition to approximately 60% (at 100 ⁇ M). In other words, KAVA-3 and KAVA-2 exerted a strong TNF- ⁇ production inhibitory effect, and KAVA-5 exerted a TNF- ⁇ inhibitory effect to an extent similar to that of KAVA-1.
  • mice of 6 weeks age Male BALB/cAnNCrj mice of 6 weeks age were purchased from Japan Charles River, and those having body weights of 30 g or lower were tested.
  • mice groups each group consisting of six mice, were provided; i.e., 1) a group to which distilled water for injection was administered (non-treated group) (N); 2) a group to which a 0.3% carboxymethyl cellulose-Na (0.3% CMC-Na) suspension was administered (control group) (C); and 3) five groups to which KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5, respectively, were administered (0.3% CMC-Na was used as a solvent).
  • Each of KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 was prepared to a drug liquid of 40 mg/10 ml.
  • the liquid was intraperitoneally administered at 10 ml/kg (dose: 40 mg/kg), and 0.3% CMC-Na was intraperitoneally administered at 10 ml/kg to each corresponding group.
  • Lipo-polysaccharide (LPS) product of SIGMA
  • LPS lipo-polysaccharide
  • the TNF- ⁇ production inhibitor of the present invention and the preventive, ameliorating, or therapeutic agent of the present invention for a variety of diseases caused by abnormal production of TNF- ⁇ are highly safe and exhibit an excellent effect of inhibiting TNF- ⁇ production
  • the inhibitor and agent are useful as a preventive, ameliorating, or therapeutic agent for mammals, including humans, and for a variety of diseases caused by abnormal production of TNF- ⁇ , including cachexia attributed to cancer or infectious diseases, septic shock, chronic rheumatoid arthritis, inflammatory diseases such as ulcerative colitis and Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus (SLE), rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes.

Abstract

The present invention provides a TNF-α production inhibitor containing a kavalactone as an active ingredient, which inhibitor has high safety, exerts an excellent effect of inhibiting TNF-α production, and is useful as a drug or an animal drug for preventing, ameliorating, or treating diseases such as cachexia attributed to cancer or infectious diseases, chronic rheumatoid arthritis, inflammatory diseases, osteoarthritis, systemic lupus erythematosus (SLE), rejection during bone marrow transplantation, multiple organ failure, AIDS, meningitis, hepatitis, and type-II diabetes. The present invention also provides a preventive, ameliorating, or therapeutic agent for diseases caused by abnormal production of TNF-α, the agent containing a kavalactone as an active ingredient.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention relates to a TNF-α production inhibitor containing a kavalactone as an active ingredient, and to a preventive, ameliorating, or therapeutic agent for diseases caused by abnormal production of TNF-α.
  • 2. Background Art
  • TNF (Tumor Necrosis Factor) was discovered as an antitumor cytokine, and has been elucidated to have carcinostatic activity (i.e., the effect of inhibiting cancer cell growth or necrotizing cancer cells), and to participate in a series of inflammatory responses or immunoreactions, as well as in differentiation or maturation of cells.
  • Recent studies have shown that excessive production of TNF-α induces onset of a variety of diseases, including cachexia attributed to cancer or infectious diseases (Nature, 316: 552, 1985), septic shock (J. Immunol., 145: 4185, 1990; Science, 229: 869, 1985; Shock, 30: 1990), chronic rheumatoid arthritis (Ann. Rheum. Dis., 49: 665, 1990; Lancet, 344: 1105, 1994; Lancet, 344: 1125, 1994; British J. Rheum., 34: 334, 1995), inflammatory diseases such as ulcerative colitis and Crohn disease (Arch. Dis. Child, 66: 561, 1991; Gastroenterology), osteoarthritis (Arthritis Rheum., 36: 819, 1993), Kawasaki's disease (Clin. Immunol. Immunopathol., 56: 29, 1990), multiple sclerosis (N. Engl. J. Med., 325(7): 467, 1991), Behchet's disease (J. Rheumatol., 17: 1107, 1990), systemic lupus erythematosus (SLE) (Arthritis Rheum., 32: 146, 1989), rejection during bone marrow transplantation (J. Exp. Med., 175: 405, 1992), multiple organ failure (Rinshoi, 17(20), 2006, 1991), malaria (Science, 237: 1210, 1987), AIDS (J. Acquir. Immune Defic. Syndr., 5: 1099, 1992), meningitis (Lancet, 1: 355, 1987), hepatitis (Kozo Kanno, Kanzo, 33: 213, 1992), and type-II diabetes (Science, 259: 87, 1993).
  • The aforementioned diseases caused by excessive production of TNF-α have hitherto been treated from a mere palliative approach by use of steroid agents, anti-inflammatory agents, antibiotics, etc., and drugs for fundamentally treating the diseases have not yet been developed.
  • Kava is a plant found in Fiji and belongs to Piperaceae, Piper L. (nomenclature: Piper Methysticum Forst., alias: Yangona). Since anesthetic beverages are obtained from the kava root, in Oceania, kava is widely cultivated by privileged people and is used in traditional ceremonies or events (Chem. Australia. October 377-378 (1987)).
  • It has been reported that an extract obtained from the dried kava root through extraction with water contains a class of α-pyrone derivatives called kavalactones which induce numbness of the lips or tongue or exert sedative effect, such as methysticin (Chem. Australia. October 377-378 (1987), Planta Med. 64 504-506 (1998)).
  • Studies performed in the University of New South Wales have elucidated that kavalactones exert a sedative effect through a mechanism different from those of other sedative drugs which exert sedative effects when being bound to receptors present in the brain (Planta Med. 65 507-510 (1998)). It has also been reported that kavalactones exert an analgesic effect in a manner different from that of a formulated analgesic drug such as aspirin, and that, unlike morphine, kavalactones are not bound to receptors in the brain (e.g., European Patent Application Laid-Open Nos. 664131 and 523591, and Japanese Kohyo (PCT) Patent Publication No. 5-502457).
  • It has also been reported that kava extract exerts an antibacterial effect and is useful for treating Helicobacter pylori infection (German Patent Application Laid-Open No. 19716660), and that the kava extract exerts a neuroprotective effect and is useful for treating brain dysfunction, Alzheimer's disease, brain injury, etc. (e.g., European Patent Application Laid-Open No. 523591, and Japanese Kohyo (PCT) Patent Publication No. 5-502457).
  • However, until the present invention was attained, kavalactones and kava extract have not been known to exert the effect of inhibiting TNF-α production.
  • SUMMARY OF THE INVENTION
  • In view of the foregoing, the present inventors have performed studies on naturally occurring substances which inhibit production of TNF-α, and have found that kavalactones contained in kava extract exert an excellent effect of inhibiting TNF-α production, and that the kavalactones are useful as TNF-α production inhibitors and as preventive, ameliorating, or therapeutic agents for a variety of diseases caused by abnormal production of TNF-α. The present invention has been accomplished on the basis of this finding.
  • Thus, an object of the present invention is to provide a drug which is endowed with high safety, inhibits TNF-α production, and is useful as a preventive or therapeutic agent for the aforementioned diseases.
  • Accordingly, the present invention provides a TNF-α production inhibitor comprising a kavalactone as an active ingredient.
  • The present invention also provides a preventive, ameliorating, or therapeutic agent comprising a kavalactone as an active ingredient for diseases caused by abnormal production of TNF-α.
  • The present invention further provides a method for the treatment of diseases caused by abnormal production of TNF-α, which method comprises administering an effective amount of a kavalactone.
  • The present invention further provides use of a kavalactone for the manufacture of a TNF-α production inhibitor.
  • Still, the present invention provides use of a kavalactone for the manufacture of a medicament for preventing, ameliorating, or treating diseases caused by abnormal production of TNF-α.
  • Preferably, the kavalactone is one or more species selected from the group consisting of desmethoxyyangonin, dihydrokavain, kavain, yangonin, methysticin, dihydromethysticin, and 7,8-epoxyyangonin.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • Various other objects, features, and many of the attendant advantages of the present invention will be readily appreciated as the same becomes better understood with reference to the following detailed description of the preferred embodiments when considered in connection with accompanying drawings, in which:
  • FIG. 1 shows a graph indicating inhibitory effects of KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 on TNF-α production in BALB/3T3 cells stimulated by okadaic acid; and
  • FIG. 2 shows a graph showing investigation results of inhibitory effects of KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 on TNF-α production in serum samples, the TNF-α production having been induced by intraperitoneally administering KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 to BALB/c mice, followed by administration of LPS immediately thereafter.
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • Kavalactone contained, as an active ingredient, in the TNF-α production inhibitor of the present invention refers to a class of α-pyrone derivatives contained in the root of kava (Piper Methysticum G. Forst) which belongs to Piperaceae, Piper L. Specific examples of the α-pyrone derivatives include desmethoxyyangonin, dihydrokavain, kavain, yangonin, methysticin, dihydromethysticin, and 7,8-epoxyyangonin, which are represented by the following formulas.
    Figure US20060121132A1-20060608-C00001
  • These α-pyrone derivatives have a variety of isomers, including geometrical isomers such as cis-isomers and trans-isomers, optical isomers such as d-isomers and l-isomers, and rotational isomers. In the present invention, any of such isomers can be used, so long as the isomer can exert the effect of inhibiting TNF-α production. In the present invention, these pyrone derivatives also include racemic modifications and a mixture of diastereomers.
  • From the viewpoint of the effect of inhibiting TNF-α production, examples of particularly preferred kavalactones include desmethoxyyangonin (KAVA-3: compound 1), (+)-dihydrokavain (KAVA-4: compound 2), (+)-kavain (KAVA-5: compound 3), yangonin (KAVA-2: compound 4), (+)-methysticin (KAVA-1: compound 5), (+)-dihydromethysticin (KAVA-6: compound 6), and 7,8-epoxyyangonin (compound 7).
  • These α-pyrone derivatives may be used singly or in combination of two or more species as kavalactone employed in the TNF-α production inhibitor of the present invention.
  • Kavalactones used in the present invention may be obtained from the kava root by means of a known extraction method, or obtained through synthesis by means of a published method (Acta Chemica Scandinavica B 30, 7: 613-678, 1976; Planta Med., 64: 504, 1998).
  • In the case in which kavalactones are obtained through extraction, the aforementioned kavalactones separated from kava extract and purified can be used. However, an extraction fraction containing a plurality of compounds may also be used, so long as the fraction exhibits the effect of inhibiting TNF-α production.
  • Since the thus-obtained kavalactones exert an excellent effect of inhibiting TNF-α production in vivo and in vitro as described below in Examples, the kavalactones can be used as a TNF-α production inhibitor for mammals including humans, and as a preventive, ameliorating, or therapeutic agent for a variety of diseases caused by abnormal production of TNF-α, including cachexia attributed to cancer or infectious diseases, septic shock, chronic rheumatoid arthritis, inflammatory diseases such as ulcerative colitis and Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus (SLE), rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes.
  • When the kavalactone according to the present invention is administered as a drug, the amount and frequency of administration vary with pathological conditions, age, weight, manner of administration, and other conditions. In the case of peroral administration, the daily dose is typically 0.1-1,000 mg for adults, but the daily dose varies with pathological conditions and other conditions (e.g., 10-500 mg or 30-300 mg). Regarding the case in which pure kavalactone is employed, a daily dose of 200 mg has been reported (Kretschmer “Kavain als Psychopharmkon,” NMW 4/1970, 154-158).
  • When the kavalactone according to the present invention is used as a TNF-α production inhibitor and as a preventive, ameliorating, or therapeutic agent for diseases caused by abnormal production of TNF-α, the kavalactone is prepared in the form of a typical pharmaceutical product. For example, the kavalactone is formulated in a form suitable for oral administration or parenteral administration (e.g., intraarticular administration or enteric administration), such as a pharmaceutical composition obtained by mixing the drug of the present invention with a pharmaceutically acceptable carrier (e.g., an excipient, a binder, a disintegrant, a sweetening agent, a flavoring agent, an emulsifying agent, a diluent, or a dissolution promoter) and processing to have a product form of, for example, tablet, pill, powder, granule, capsule, troche, syrup, solution, emulsion, suspension, or injection.
  • Examples of excipients include lactose, cornstarch sucrose, glucose, sorbitol, and crystalline cellulose. Examples of binders include polyvinyl alcohol, polyvinyl ether, ethyl cellulose, methyl cellulose, gum arabi, tragacanth, gelatin, shellac, hydroxypropyl cellulose, hydroxypropyl starch, and polyvinyl pyrrolidone.
  • Examples of disintegrants include starch, agar, gelatin powder, crystalline cellulose, calcium carbonate, sodium hydrogencarbonate, calcium citrate, dextran, and pectin. Examples of lubricants include magnesium stearate, talc, polyethylene glycol, silica, and hydrogenated vegetable oil. Any pharmaceutically acceptable coloring agent may be used. Examples of sweetening and flavoring agents include cocoa powder, menthol, aromatic acids, peppermint oil, borneol, and cinnamon powder. If necessary, tablets or granules may optionally be subjected to sugar coating, gelatin coating, or similar coating.
  • In the case of preparation of injections, if necessary, a pH-regulating agent, a buffer, a stabilizer, or a preservative is added to the injections, to thereby prepare agents for subcutaneous injection, intramuscular injection, or intravenous injection, by means of a customary method. Injection preparations may be stored in a container and freeze-dried, to thereby provide solid products, and the solid products may be prepared into injections upon use. A single dose of the injection may be stored in a container, or a plurality of doses may be stored in a single container.
  • EXAMPLES
  • The present invention will next be described in more detail by way of examples, which should not be construed as limiting the invention thereto.
  • Example 1 Isolation of Components having a TNF-α Release Inhibitory Effect through Extraction
  • Dried kava root (1.0 kg) was subjected to extraction by use of methanol for 16 days. The resultant mixture was subjected to filtration under reduced pressure, and the filtrate was concentrated under reduced pressure, to thereby yield a residue (152.0 g). The entirety of the residue was partitioned by use of water and ethyl acetate (AcOEt), and the resultant ethyl acetate phase was concentrated under reduced pressure, to thereby yield an ethyl acetate extract (76.17 g). The entirety of the extract was added to a silica gel column (product of Merck, 1 kg (70-230 mesh 500 g+230-400 mesh 500 g)) for chromatography by use of n-hexane/ethyl acetate. In the course of chromatography, the ethyl acetate concentration of the solvent was gradually elevated so as to effect elution/extraction in the following concentration profile: n-hexane (500 ml); 5% ethyl acetate/n-hexane (500 ml); 10% ethyl acetate/n-hexane (500 ml); 15% ethyl acetate/n-hexane (500 ml); 20% ethyl acetate/n-hexane (500 ml); 25% ethyl acetate/n-hexane (500 ml); 30% ethyl acetate/n-hexane (500 ml); 35% ethyl acetate/n-hexane (500 ml); 40% ethyl acetate/n-hexane (500 ml); 45% ethyl acetate/n-hexane (500 ml); 50% ethyl acetate/n-hexane (500 ml); 60% ethyl acetate/n-hexane (500 ml); 70% ethyl acetate/n-hexane (500 ml); 80% ethyl acetate/n-hexane (500 ml); 85% ethyl acetate/n-hexane (500 ml); 90% ethyl acetate/n-hexane (500 ml); and ethyl acetate (500 ml). Collection of fractions was initiated from the eluate corresponding to 20% ethyl acetate/n-hexane, with the volume of each fraction being 20 ml. The eluate corresponding to fractions 65 to 74 was concentrated under reduced pressure, to thereby yield 1.48 g of crude crystals. The crystals were recrystallized from ethyl acetate/n-hexane, to thereby yield 1.218 g of compound 1: 5,6-dehydrokavain, as pale yellow needles. In a similar manner, crude crystals (3.78 g) obtained from the eluate corresponding to fractions 75 to 82 were recrystallized from ethyl acetate/n-hexane, to thereby yield 3.513 g of compound 2: dihydrokavain, as colorless needles. Crude crystals (5.36 g) obtained from the eluate corresponding to fractions 86 to 94 were recrystallized from ethyl acetate/ether, to thereby yield 4.744 g of compound 3: kavain, as colorless prisms. Crude crystals (1.27 g) obtained from the eluate corresponding to fractions 97 to 102 were recrystallized from ethyl acetate/ether, to thereby yield 0.847 g of compound 4: yangonin, as pale yellow prisms. The eluent corresponding to fractions 103 to 115 was concentrated under reduced pressure, to thereby yield a residue (8.21 g). The residue was subjected to chromatography by use of a silica gel column (product of Merck, 300 g (70-230 mesh 150 g+230-400 mesh 150 g)) and chloroform/ether solvent. In the course of chromatography, the ether concentration was gradually elevated. From the eluent corresponding to 25% ether/chloroform, 1.322 g of compound 4: yangonin was obtained. Subsequently, crude crystals obtained from the eluent corresponding to 30% ether/chloroform was recrystallized from ethyl acetate/ether, to thereby yield 5.661 g of compound 5: methysticin, as colorless needles. A residue (385 mg) obtained from the eluent corresponding to fractions 138 to 142 was placed on a Sephadex LH-20 column (product of Pharmacia, 30 g), and chromatography was performed by use of chloroform/methanol (1:1) solvent for development. Twenty mL of the eluent was collected per fraction (Fr.). From the eluent corresponding to fractions 5 to 7, 59 mg of pale yellow-white novel compound 7: 7,8-epoxyyangonin was obtained.
  • Compound 1 (KAVA-3) 5,6-Dehydrokavain (Desmethoxyyangonin); (4-methoxy-6-(2-phenylvinyl-2H-pyran-2-one):
  • m.p. 138-140° C., pale yellow needles
  • EI-MS: m/z 228 (M+, 100%), 211 (10%), 200 (36%), 185 (15%), 157 (27%)
  • HR-MS: m/z 228.0763, C14H12O3 requires 228.0787, FT-IR (KBr) νmax cm−1: 3081, 1721 (C═O), 1644 (C═C), 1611, 1557, 1256, 1154, UV (EtOH) λmax nm (log ε): 344.5 (4.32), 255 (4.05), 231.5 (4.15), 225 (4.14), 209 (4.28)
  • 600 MHz 1H NMR (CDCl3): δ 3.82 (3H, s, 4-OMe), 5.50 (1H, d, J=2.2 Hz, H-3), 5.95 (1H, d, J=2.2 Hz, H-5), 6.58 (1H, d, J=16.2 Hz, H-7), 7.33 (1H, br. t, J=7.1 Hz, 4′-H), 7.38 (2H, br. t, J=7.1 Hz, 3′,5′-H), 7.49 (2H, br. d, J=7.1 Hz, 2′, 6′-H), 7.50 (1H, d, J=16.2 Hz, H-8). 150 MHz 13C NMR (CDCl3): δ 55.9 (q, 4-OMe), 88.8 (d, C-3), 101.3 (d, C-5), 118.6 (d, C-7), 127.4 (d, C-2′ and C-6′), 128.9 (d, C-3′ and C-5′), 129.4 (d, C-4′), 135.2 (s, C-1′), 135.7 (d, C-8), 158.6 (s, C-6), 164.0 (s, C-2), 171.0 (s, C-4).
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00002
  • Compound 2 (KAVA-4) (+)-Dihydrokavain; (4-methoxy-2-(2-phenylvinyl)-2H, 3H-oxin-2-one
  • m.p. 57-60° C., colorless needles
  • [α]D 20+45.7°(c0.50, CHCl3)
  • EI-MS: m/z 232 (M+, 61%), 200 (30%), 173 (15%), 141 (26%), 127 (100%)
  • HR-MS: m/z 232.1094, C14H16O3 requires 232.1100
  • FT-IR (KBr) νmax cm−1: 1707 (C═O), 1624, 1225, 1090, 1038
  • UV (EtOH) λmax nm (log s): 233.0 (4.01), 207.0 (4.08)
  • 600 MHz 1H NMR (CDCl3): δ 1.93 (1H, m, H-7), 2.13 (1H, m, H-7), 2.30 (1H, dd, J=3.8, 17.0 Hz, H-5), 2.50 (1H, ddd, J=1.6, 11.8, 17.0 Hz, H-5), 2.79 (1H, m, H-8), 2.88 (1H, m, H-8), 3.72 (3H, s, 4-OMe), 4.36 (1H, m, H-6), 5.14 (1H, d, J=1.6 Hz, H-3), 7.20 (1H, br. t, J=7.1 Hz, 4′-H), 7.21 (2H, br. d, J=7.1 Hz, 2′,6′-H), 7.49 (2H, br. t, J=7.1 Hz, 3′, 5′-H).
  • 150 MHz 13C NMR (CDCl3) δ 30.9 (t, C-8), 33.0 (t, C-5), 36.3 (t, C-7), 55.9 (q, 4-OMe), 74.7 (d, C-6), 90.3 (d, C-3), 126.1 (d, C-4′), 128.4 (d, C-2′ and C-6′), 128.5 (d, C-3′ and C-5′), 140.8 (s, C-1′), 167.2 (s, C-2), 172.7 (s, C-4).
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00003
  • Compound 3 (KAVA-5) (+)-Kavain; (4-methoxy-2-(2-phenylvinyl)-2H,3H-oxin-2-one)
  • m.p. 106-108° C., colorless needles
  • [α]DD 20+116.3°(c1.01, CHCl3)
  • EI-MS: m/z 230 (M+, 27%), 202 (43%), 186 (13%), 128 (31%), 98 (100%)
  • HR-MS: m/z 230.0951, C14H14O3 requires 230.0943
  • FT-IR (KBr) νmax cm−1: 1703 (C═O), 1626, 1248, 1231
  • UV (EtOH) λmax nm (log ε): 244.5 (4.41), 205.0 (4.48)
  • 600 MHz 1H NMR (CDCl3): δ 2.54 (1H, dd, J=4.4, 17.0 Hz, H-5), 2.66 (1H, ddd, J=1.4, 11.0, 17.0 Hz, H-5), 3.76 (3H, S, 4-OMe), 5.05 (1H, ddd, J=4.4, 6.3, 11.0 Hz, H-6), 5.19 (1H, d, J=1.4 Hz, H-3), 6.26 (1H, dd, J=6.3, 15.9 Hz, H-7), 6.73 (1H, br, d, J=15.9 Hz, H-8), 7.27 (1H, br. t, J=7.1, Hz, 4′-H), 7.33 (2H, br. t, J=7.1 Hz, 3′,5′-H), 7.39 (2H, br. d, J=7.1 Hz, 2′,6′-H).
  • 150 MHz 13C NMR (CDCl3): δ 33.2 (t, C-5), 56.0 (q, 4-OMe), 75.8 (d, C-6), 90.4 (d, C-3), 125.4 (d, C-7), 126.6 (d, C-2′ and C-6′), 128.2 (d, C-4′), 128.6 (d, C-3′ and C-5′), 133.0 (d, C-8), 135.7 (s, C-1′), 166.6 (s, C-2), 172.2 (s, C-4).
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00004
  • Compound 4 (KAVA-2) Yangonin; (4-methoxy-6-(2-(4-methoxyphenyl)vinyl)-2H-pyran-2-one)
  • m.p. 153-155° C., yellow needles
  • EI-MS: m/z 258 (M+, 100%), 230 (38%), 215 (13%), 187 (33%)
  • HR-MS: m/z 258.0896, C15H14O4 requires 258.0892
  • FT-IR (KBr) νmax cm−1: 1717 (C═O), 1644 (C═C), 1603, 1555, 1256, 1154
  • UV (EtOH) λmax nm (log ε): 357.5 (4.42), 260.0 (3.89), 218.0 (4.28)
  • 600 MHz 1H NMR (CDCl3): δ 3.81 (3H, s, 4′-OMe), 3.82 (3H, s, 4-OMe), 5.47 (1H, d, J=2.2 Hz, H-3), 5.89 (1H, d, J=2.2 Hz, H-5), 6.44 (1H, d, J=15.7 Hz, H-7), 6.90 (2H, d, J=8.8 Hz, 3′,5′-H), 7.44 (2H, d, J=8.8 Hz, 2′,6′-H), 7.45 (1H, d, J=15.7 Hz, H-8)
  • 150 MHz 13C NMR (CDCl3): δ 55.3 (q, 4′-OMe), 55.8 (q, 4-OMe), 88.3 (d, C-3), 100.4 (d, C-5), 114.3 (d, C-3′ and C-5′), 116.3 (d, C-7), 127.9 (s, C-1′), 128.9 (d, C-2′ and C-6′), 135.4 (d, C-8), 159.0 (s, C-6), 160.7 (s, C-4′), 164.1 (s, C-2), 171.2 (s, C-4).
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00005
  • Compound 5 (KAVA-1) (+)-Methysticin; (2-(2-benzo[3,4-d]1,3-dioxolan-5-ylvinyl-4-methoxy-2H,3H,-oxin-2-one) m.p. 139-141° C., colorless needles
  • [α]DD 20+115.9°(c0.50, CHCl3)
  • EI-MS: m/z 274 (M+, 100%), 246 (10%), 175 (19%), 148 (81%), 135 (82%)
  • HR-MS: m/z 274.0833, C15H14O5 requires 274.0841
  • FT-IR (KBr) νmax cm−1: 1711 (C═O), 1628, 1252, 1217, 1038
  • UV (EtOH) λmax nm (log ε): 305.5 (3.89), 264.5 (4.13), 225.5 (4.38), 207.0 (4.44)
  • 600 MHz 1H NMR (CDCl3): δ 2.53 (1H, dd, J=4.4, 17.0 Hz, H-5), 2.65 (1H, ddd, J=1.4, 11.0, 17.0 Hz, H-5), 3.77 (3H, s, 4-OMe), 5.02 (1H, ddd, J=4.4, 6.6, 11.0 Hz, H-6), 5.19 (1H, d, J=1.4 Hz, H-3), 6.07 (2H, S, —O—CH2—O—), 6.09 (1H, dd. J=6.6, 15.9 Hz, H-7), 6.64 (1H, br. d, J=15.9 Hz, H-8), 6.76 (1H, d, J=8.0 Hz, 5′-H), 6.83 (1H, dd, J=1.9, 8.8 Hz, 6′-H), 6.92 (1H, d, J=1.9 Hz, 2′-H).
  • 150 MHz 13C NMR (CDCl3): δ 33.3 (t, C-5), 56.1 (q, 4-OMe), 76.0 (d, C-6), 90.5 (d, C-3), 101.2 (t, —O—CH2—O—), 105.8 (d, C-2′), 108.3 (d, C-5′), 121.7 (d, C-6′), 123.6 (d, C-7), 130.1 (s, C-1′), 132.9 (d, C-8), 147.8 (s, C-4′), 148.1 (s, C-3′), 166.8 (s, C-2), 172.3 (s, C-4).
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00006
  • Compound 6 (KAVA-6) (+)-Dihydromethysticin (Planta Med., 64, 504-506, 1998)
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00007
  • Compound 7 7,8-Epoxyyangonin; (4-Methoxy 6-[2-(4-methoxyphenyl) oxirane]-2H-pyran-2-one)
  • [α]DD 18+13.04° (c1.51, CHCl3)
  • EI-MS: m/z 274 (M+, 5%), 258 (100%), 230 (64%), 187 (66%)
  • HR-MS: m/z 274.0851, C15H14O5 requires 274.0841
  • FT-IR (KBr) νmax cm−1: 2940, 1721 (C═O), 1645 (C═C), 1613, 1566, 1252, (C—O—C), 1181
  • 600 MHz 1H NMR (CDCl3): δ 3.69(3H, s, 4′-OMe), 3.77(3H, s, 4-OMe), 5.22(1H, d, J=2.2 Hz, H-3), 5.73(1H, d, J=2.2 Hz, H-5), 6.82(2H, d, J=8.7 Hz, 2′,6′-H), 7.25(2H, d, J=8.7 Hz, 3′,5′-H).
  • 150 MHz 13C NMR(CDCl3): δ 42.9(d, C-7), 45.5(d, C-8), 55.1 (q, 4′-OMe), 55.6(q, 4-OMe), 87.6(d, C-3), 101.2(d, C-5), 113.8(d, C-3′ and C-5′), 128.4(d, C-2′ and C-6′), 129.4(s, C-1′), 158.5(s, C-4′), 162.9(s, C-6), 164.1(s, C-2), 107.5(s, C-4).
  • The structural formula thereof is shown below.
    Figure US20060121132A1-20060608-C00008
  • Test Examples in relation to the present invention will next be described.
  • Test Example 1 Inhibition of TNF-α Production (In Vitro)
  • In response to stimulation by okadaic acid (9,10-deepithio-9,10-didehydroacanthifolicin), BALB/3T3 cells produce TNF-α. The compounds of the present invention were investigated in terms of inhibitory effect on TNF-α production.
  • An MEM medium (product of Nissui) containing 10% fetal calf serum (product of Biocell Laboratory) was injected into 12-well multiplates (product of Corning), and BALB/3T3 cells were disseminated at 2×105 cells/well. The cells were cultured in a carbon dioxide gas incubator (5% CO2, humidified, 37° C.). Subsequently, KAVA-1, KAVA-2, KAVA-3, KAVA-4, or KAVA-5 was added to the wells at a concentration shown in FIG. 1, and the cells were cultured for one hour. No KAVA compound was added to the control wells. After completion of culturing, okadaic acid (carcinogenisis promoter isolated from Halichondria okadai) was added to each well at a final concentration of 0.2 μM, and culturing was performed for 24 hours. After completion of this culturing, the TNF-α concentration of the supernatant of each well was measured by means of ELISA system (product of Genzyme). The results are shown in FIG. 1. In FIG. 1, the amount of TNF-α release corresponding to each compound concentration is represented by a percent concentration based on the amount of TNF-α release measured for the control (100%).
  • KAVA-3 was found to inhibit TNF-α production to approximately 60% (at 10 μM) the TNF-α production of the control; to approximately 22% (at 50 μM) the TNF-α production of the control; and to approximately 0% (complete inhibition) (at 100 μM). KAVA-2 was found to inhibit TNF-α production to approximately 22% (at 100 μM). KAVA-5 and KAVA-1 exerted similar inhibitory effects; i.e., exerted inhibition to approximately 39% (at 100 μM). KAVA-4 exerted no inhibitory effect at concentrations of 50 μM or less, but exerted inhibition to approximately 60% (at 100 μM). In other words, KAVA-3 and KAVA-2 exerted a strong TNF-α production inhibitory effect, and KAVA-5 exerted a TNF-α inhibitory effect to an extent similar to that of KAVA-1.
  • Test Example 2 TNF-α inhibitory effect (in vivo)
  • Male BALB/cAnNCrj mice of 6 weeks age were purchased from Japan Charles River, and those having body weights of 30 g or lower were tested.
  • Seven mice groups, each group consisting of six mice, were provided; i.e., 1) a group to which distilled water for injection was administered (non-treated group) (N); 2) a group to which a 0.3% carboxymethyl cellulose-Na (0.3% CMC-Na) suspension was administered (control group) (C); and 3) five groups to which KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5, respectively, were administered (0.3% CMC-Na was used as a solvent).
  • Each of KAVA-1, KAVA-2, KAVA-3, KAVA-4, and KAVA-5 was prepared to a drug liquid of 40 mg/10 ml. The liquid was intraperitoneally administered at 10 ml/kg (dose: 40 mg/kg), and 0.3% CMC-Na was intraperitoneally administered at 10 ml/kg to each corresponding group. Lipo-polysaccharide (LPS) (product of SIGMA) was dissolved in physiological saline, and the solution was intraperitoneally administered to each mouse in an amount of 0.2 ml (50 μg/mouse) immediately after administration of the drug liquid.
  • After 90 minutes from administration of LPS, blood was collected from the eye socket, and the collected sample was allowed to stand for one hour at room temperature. Subsequently, the sample was centrifuged at 11,000 rpm for five minutes, and the serum was collected. The TNF-α in the serum was assayed by use of an ELISA kit ([(m) TNFα] mouse ELISA system, product of Amersham Pharmacia Biotech K.K.). The results are shown in FIG. 2.
  • The results shown in FIG. 2 indicate that KAVA-4 significantly (p<0.5) inhibited TNF-α production as compared with the control group (C), and the TNF-α production inhibitory effect exerted by KAVA-1 was almost comparable to that exerted by KAVA-2.
  • As is clear from the above test results, all kavalactones of the present invention (compounds 1 to 5) can inhibit TNF-α production. The TNF-α production inhibitory effect of compounds 6 and 7 can also be confirmed on the basis of the above test results.
  • Since the TNF-α production inhibitor of the present invention and the preventive, ameliorating, or therapeutic agent of the present invention for a variety of diseases caused by abnormal production of TNF-α are highly safe and exhibit an excellent effect of inhibiting TNF-α production, the inhibitor and agent are useful as a preventive, ameliorating, or therapeutic agent for mammals, including humans, and for a variety of diseases caused by abnormal production of TNF-α, including cachexia attributed to cancer or infectious diseases, septic shock, chronic rheumatoid arthritis, inflammatory diseases such as ulcerative colitis and Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus (SLE), rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes.

Claims (7)

1-10. (canceled)
11. A method of treating one or more diseases caused by abnormal production of TNF-α in a patient in need thereof, wherein said disease is selected from the group consisting of cachexia related to infectious disease, septic shock, chronic rheumatoid arthritis, ulcerative colitis, Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus, rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes, comprising administering to said patient in need thereof an effective dose methysticin and at least one additional kavalactone.
12. The method of claim 11, wherein said additional kavalactone is selected from the group consisting of desmethoxyyangonin, dihydrokavain, kavain, yangonin, dihydromethysticin, and 7,8-epoxyyangonin.
13. A method of treating one or more diseases caused by abnormal production of TNF-α in a patient in need thereof, wherein said disease is selected from the group consisting of cachexia related to infectious disease, septic shock, chronic rheumatoid arthritis, ulcerative colitis, Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus, rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes, comprising administering to said patient in need thereof an effective dose of yangonin and at least one additional kavalactone.
14. The method of claim 13, wherein said additional kavalactone is selected from the group consisting of desmethoxyyangonin, dihydrokavain, kavain, methysticin, dihydromethysticin, and 7,8-epoxyyangonin.
15. A method of treating one or more diseases caused by abnormal production of TNF-α in a patient in need thereof, wherein said disease is selected from the group consisting of cachexia related to infectious disease, septic shock, chronic rheumatoid arthritis, ulcerative colitis, Crohn disease, osteoarthritis, Kawasaki's disease, multiple sclerosis, Behchet's disease, systemic lupus erythematosus, rejection during bone marrow transplantation, multiple organ failure, malaria, AIDS, meningitis, hepatitis, and type-II diabetes, comprising administering to said patient in need thereof an effective dose of dihydrokavain and at least one additional kavalactone.
16. The method of claim 15, wherein said additional kavalactone is selected from the group consisting of desmethoxyyangonin, kavain, yangonin, methysticin, dihydromethysticin.
US11/335,641 2000-12-18 2006-01-20 TNF-alpha production inhibitor comprising kavalactone as an active ingredient Abandoned US20060121132A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/335,641 US20060121132A1 (en) 2000-12-18 2006-01-20 TNF-alpha production inhibitor comprising kavalactone as an active ingredient

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2000-383095 2000-12-18
JP2000383095A JP2001316260A (en) 2000-02-29 2000-12-18 TNF-alpha PRODUCTION INHIBITOR CONTAINING KAVALACTONES AS ACTIVE INGREDIENT
US09/941,713 US6608105B2 (en) 2000-12-18 2001-08-30 TNF-α production inhibitor comprising kavalactone as an active ingredient
US10/185,045 US7199152B2 (en) 2000-12-18 2002-07-01 TNF-α production inhibitor comprising kavalactone as an active ingredient
US11/335,641 US20060121132A1 (en) 2000-12-18 2006-01-20 TNF-alpha production inhibitor comprising kavalactone as an active ingredient

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/185,045 Division US7199152B2 (en) 2000-12-18 2002-07-01 TNF-α production inhibitor comprising kavalactone as an active ingredient

Publications (1)

Publication Number Publication Date
US20060121132A1 true US20060121132A1 (en) 2006-06-08

Family

ID=18850805

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/941,713 Expired - Fee Related US6608105B2 (en) 2000-12-18 2001-08-30 TNF-α production inhibitor comprising kavalactone as an active ingredient
US10/185,045 Expired - Fee Related US7199152B2 (en) 2000-12-18 2002-07-01 TNF-α production inhibitor comprising kavalactone as an active ingredient
US11/335,641 Abandoned US20060121132A1 (en) 2000-12-18 2006-01-20 TNF-alpha production inhibitor comprising kavalactone as an active ingredient

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/941,713 Expired - Fee Related US6608105B2 (en) 2000-12-18 2001-08-30 TNF-α production inhibitor comprising kavalactone as an active ingredient
US10/185,045 Expired - Fee Related US7199152B2 (en) 2000-12-18 2002-07-01 TNF-α production inhibitor comprising kavalactone as an active ingredient

Country Status (5)

Country Link
US (3) US6608105B2 (en)
EP (1) EP1222925B1 (en)
AT (1) ATE289511T1 (en)
CA (1) CA2356812A1 (en)
DE (1) DE60109018T2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009110947A2 (en) * 2007-12-19 2009-09-11 Alba Therapeutics Corporation Compositions and methods for inhibiting cytokine production
WO2016179587A1 (en) * 2015-05-07 2016-11-10 Kuality Herbceutics Llc Therapeutic compounds and methods of use thereof
US10457654B2 (en) 2016-10-17 2019-10-29 Genentech, Inc. Therapeutic compounds and methods of use thereof
US10624943B2 (en) 2013-11-11 2020-04-21 Kuality Herbceutics, LLC Kava derived therapeutic compounds and methods of use thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6608105B2 (en) * 2000-12-18 2003-08-19 Meiji Dairies Corporation TNF-α production inhibitor comprising kavalactone as an active ingredient
CA2451133C (en) * 2001-05-08 2012-09-18 Toray Industries, Inc. Therapeutic agent for sepsis
US20030229137A1 (en) * 2001-10-12 2003-12-11 Shoujun Chen Methods of inhibiting osteoclast activity
CA2525012A1 (en) * 2003-05-08 2004-12-02 Wyeth Protein kinase c zeta as a drug target for arthritis and other inflammatory diseases
CN107090023B (en) * 2017-03-23 2020-06-05 桂林八加一药业股份有限公司 Selective TNFR1 antagonistic peptide SN10 and application thereof in rheumatoid arthritis

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5296224A (en) * 1990-09-12 1994-03-22 Dr. Wilmar Schwabe Gmbh & Co. Kava-kava extract, process for the production thereof and use thereof
US6143300A (en) * 1998-08-11 2000-11-07 Robin Ann Stevenot Fem-Ease, a supplement for the symptoms of cystitis, urinary tract infections and premenstrual syndrome
US6288109B1 (en) * 1997-11-14 2001-09-11 Dr. Willmar Schwabe Gmbh & Co. Pyranones, method for the production and use thereof
US20010031783A1 (en) * 2000-03-03 2001-10-18 Steiner Gregory Gene Alpha-pyrone compositions and method for the chemoprevention of cancer
US20020002146A1 (en) * 2000-02-11 2002-01-03 Halevie-Goldman Brian D. Compositions and methods for the production of S-adenosylmethionine within the body
US20020077351A1 (en) * 2000-12-18 2002-06-20 Meiji Dairies Corporation TNF-alpha production inhibitor comprising kavalactone as an active ingredient

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US77351A (en) * 1868-04-28 Impeovement in cobsr-plafrebs
US173540A (en) * 1876-02-15 Improvement in air-guns
DE1617812A1 (en) * 1967-06-05 1972-02-17 Spezialchemie Gmbh & Co Process for the production of endoanaesthetic solutions of active ingredients in kava
DE4028945A1 (en) 1990-09-12 1992-03-19 Schwabe Willmar Gmbh & Co KAWA-KAWA EXTRACT, METHOD FOR PRODUCING IT AND ITS USE
DE4123427A1 (en) 1991-07-15 1993-01-21 Schwabe Willmar Gmbh & Co USE OF A KAVA EXTRACT AND ITS INGREDIENTS
DE4401646A1 (en) 1994-01-21 1995-07-27 Krewel Werke Gmbh Optimally releasing kava extracts
DE19716660C2 (en) 1997-04-22 2002-11-14 Schwabe Willmar Gmbh & Co Preparations, in particular pharmaceutical and pharmaceutical forms based on plants, for combating Helicobacter pylori infections
US5981496A (en) * 1997-09-19 1999-11-09 Millennium Pharmaceutical, Inc. α-pyrones for treating α-pyrone responsive states
US6025363A (en) * 1998-11-17 2000-02-15 Giles, Jr.; James A. Composition for suppressing appetite
US5977120A (en) * 1998-11-17 1999-11-02 Giles, Jr.; James A. Composition for achieving an alert, yet calm state

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5296224A (en) * 1990-09-12 1994-03-22 Dr. Wilmar Schwabe Gmbh & Co. Kava-kava extract, process for the production thereof and use thereof
US6288109B1 (en) * 1997-11-14 2001-09-11 Dr. Willmar Schwabe Gmbh & Co. Pyranones, method for the production and use thereof
US6143300A (en) * 1998-08-11 2000-11-07 Robin Ann Stevenot Fem-Ease, a supplement for the symptoms of cystitis, urinary tract infections and premenstrual syndrome
US20020002146A1 (en) * 2000-02-11 2002-01-03 Halevie-Goldman Brian D. Compositions and methods for the production of S-adenosylmethionine within the body
US20010031783A1 (en) * 2000-03-03 2001-10-18 Steiner Gregory Gene Alpha-pyrone compositions and method for the chemoprevention of cancer
US20020077351A1 (en) * 2000-12-18 2002-06-20 Meiji Dairies Corporation TNF-alpha production inhibitor comprising kavalactone as an active ingredient
US20020173540A1 (en) * 2000-12-18 2002-11-21 Meiji Dairies Corporation TNF-alpha production inhibitor comprising kavalactone as an active ingredient
US6608105B2 (en) * 2000-12-18 2003-08-19 Meiji Dairies Corporation TNF-α production inhibitor comprising kavalactone as an active ingredient
US7199152B2 (en) * 2000-12-18 2007-04-03 Meiji Dairies Corporation TNF-α production inhibitor comprising kavalactone as an active ingredient

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009110947A2 (en) * 2007-12-19 2009-09-11 Alba Therapeutics Corporation Compositions and methods for inhibiting cytokine production
WO2009110947A3 (en) * 2007-12-19 2010-03-18 Alba Therapeutics Corporation Compositions and methods for inhibiting cytokine production
US10624943B2 (en) 2013-11-11 2020-04-21 Kuality Herbceutics, LLC Kava derived therapeutic compounds and methods of use thereof
US10918687B2 (en) 2013-11-11 2021-02-16 Kuality Herbceutics Llc Kava derived therapeutic compounds and methods of use thereof
US20210228672A1 (en) * 2013-11-11 2021-07-29 Kuality Herbceutics Llc Kava derived therapeutic compounds and methods of use thereof
WO2016179587A1 (en) * 2015-05-07 2016-11-10 Kuality Herbceutics Llc Therapeutic compounds and methods of use thereof
CN108026084A (en) * 2015-05-07 2018-05-11 夸利蒂赫布丘蒂克斯公司 Therapeutic compounds and its application method
US10584108B2 (en) 2015-05-07 2020-03-10 Kuality Herbceutics Llc Therapeutic compounds and methods of use thereof
US10457654B2 (en) 2016-10-17 2019-10-29 Genentech, Inc. Therapeutic compounds and methods of use thereof

Also Published As

Publication number Publication date
EP1222925B1 (en) 2005-02-23
US7199152B2 (en) 2007-04-03
US20020173540A1 (en) 2002-11-21
DE60109018D1 (en) 2005-03-31
US20020077351A1 (en) 2002-06-20
CA2356812A1 (en) 2002-06-18
US6608105B2 (en) 2003-08-19
ATE289511T1 (en) 2005-03-15
EP1222925A3 (en) 2002-09-18
EP1222925A2 (en) 2002-07-17
DE60109018T2 (en) 2005-07-21

Similar Documents

Publication Publication Date Title
US20060121132A1 (en) TNF-alpha production inhibitor comprising kavalactone as an active ingredient
AU767241B2 (en) Immunosuppressive agents
US20120041062A1 (en) Compound of salvianolic acid l, preparation method and use thereof
US8829167B2 (en) Foliamangiferosides, preparation method and use thereof
CN102526165A (en) Rhodiola effective fractions, preparation method, drug composition and uses thereof
KR101449796B1 (en) A Labdane-Type Diterpenes Compounds derived from Hedychium coronarium and a Use thereof
JP3128823B2 (en) Anticancer compound and method for producing the same
KR100656969B1 (en) Pharmaceutical composition and healthy food for treating of tuberculosis comprising Zanthoxyli Frutus extract
KR102175269B1 (en) A pharmaceutical composition comprising compounds isolated from Phlomoides umbrosa(Turcz.) Kamelin and Makhm for preventing or treating cancer
KR20130106599A (en) A labdane-type diterpenoids compounds derived from hedychium coronarium and a use thereof
JP2001316260A (en) TNF-alpha PRODUCTION INHIBITOR CONTAINING KAVALACTONES AS ACTIVE INGREDIENT
JP2006124296A (en) Medicinal composition for treatment of helicobacter pylori infectious disease
KR100979921B1 (en) Stereum ostrea extracts, lactone compounds isolated therefrom and antiobesity composition comprising the same
KR20060086458A (en) Liver protecting agent containing lignans originating in hongdoushan
JP2729340B2 (en) Dementia treatment
CN103845339B (en) Alisol A application in preparing Antiatherosclerosis medicine
KR100830202B1 (en) A composition comprising fraxidin isolated from fraxinus rhynchophylla for preventing and treating toxoplasmosis
US8921417B2 (en) Method of treating dyslipidemia using naturally occurring diterpene
CN109160927B (en) Novel amide compounds in moringa seeds and application thereof
KR100684377B1 (en) Novel ptp1b inhibitory compounds from torreya nucifera
KR100473045B1 (en) Pharmaceutical Composition Comprising Cumambrin A for Lowering Level of Blood Glucose
KR101119968B1 (en) ß-LACTONE-BASED COMPOUNDS OR PHARMACETICALLY ACCEPTABLE SALTS THEREOF, AND ANTIOBESITY COMPOSITION CONTANING THE SAME AS AN ACTIVE INGREDIENT
JP2006321728A (en) New anti-leishmania pharmaceutical composition
CN117447471A (en) Indole diketopiperazine compound, preparation method thereof and application thereof in preparation of osteoclast differentiation inhibitor
JPH0987193A (en) Anti-human immunodeficiency virus agent containing extract of ginger-family plant

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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