WO2006093793A2 - Compositions comprising actinidia and methods of use thereof - Google Patents

Compositions comprising actinidia and methods of use thereof Download PDF

Info

Publication number
WO2006093793A2
WO2006093793A2 PCT/US2006/006437 US2006006437W WO2006093793A2 WO 2006093793 A2 WO2006093793 A2 WO 2006093793A2 US 2006006437 W US2006006437 W US 2006006437W WO 2006093793 A2 WO2006093793 A2 WO 2006093793A2
Authority
WO
WIPO (PCT)
Prior art keywords
fruit
preparation
hardy kiwifruit
mammal
concentrate
Prior art date
Application number
PCT/US2006/006437
Other languages
English (en)
French (fr)
Other versions
WO2006093793A3 (en
WO2006093793A8 (en
Inventor
Julianne Lindemann
George E. Stagnitti
Robert H. Driver
Mark A. Braman
Nancy E. Fogg-Johnson
Sunyoung Kim
Eun-Jin Park
Bongcheol Kim
Mirim Jin
Hyung-Jin Jung
Sung-Seup Shin
Jin-Hwan Oh
Hwa-Jun Lee
Hyang Jeon
Original Assignee
Efficas, Inc.
Pangenomics Co., 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 Efficas, Inc., Pangenomics Co., Ltd. filed Critical Efficas, Inc.
Priority to BRPI0608042-1A priority Critical patent/BRPI0608042A2/pt
Priority to MX2007010408A priority patent/MX2007010408A/es
Priority to EP06735914A priority patent/EP1858535A4/en
Priority to JP2007557161A priority patent/JP2008531584A/ja
Priority to AU2006218875A priority patent/AU2006218875B2/en
Publication of WO2006093793A2 publication Critical patent/WO2006093793A2/en
Publication of WO2006093793A3 publication Critical patent/WO2006093793A3/en
Priority to US12/015,193 priority patent/US20080175888A1/en
Publication of WO2006093793A8 publication Critical patent/WO2006093793A8/en

Links

Classifications

    • 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)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • 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
    • A61P31/12Antivirals
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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

Definitions

  • compositions Comprising Actinidia and Methods of Use Thereof
  • the present invention relates to Actinidia, and particularly species of hardy kiwifruit, and various fractions and preparations thereof, as well as compositions comprising the same, all having the ability to prevent and/or treat a variety of diseases in which regulation of the immune response is effective, including both allergic and non- allergic inflammatory disease, viral infection, and cancer. Methods related to making and using these compositions are also described. BACKGROUND OF THE INVENTION
  • Diseases involving inflammation are characterized by the influx of certain cell types and mediators, the presence of which can lead to tissue damage and sometimes death. Diseases involving inflammation are particularly harmful when they afflict certain organs and systems, such as the respiratory system, which can result in obstructed breathing, hypoxemia, hypercapnia and lung tissue damage. In other diseases or conditions, the development of certain types of inflammation can be an important component in controlling the disease, such as in a viral infection, although damage to tissues in the area of infection is still a risk.
  • Allergic diseases are mediated in part by immunoglobulin E (IgE), while the type-2 T helper (Th2) cells, mast cells and eosinophils have also been shown to play important roles in the disease process (Maggi E., Immunotechnology, 3:233-244, 1998; Pawankar R., Curr. Opin. Allergy CHn. Immunol., 1:3-6, 2001; Vercelli D., Clin. Allergy Immunol., 16:179-196, 2002).
  • IgE immunoglobulin E
  • Th2 T helper cells type-2 T helper cells
  • mast cells eosinophils
  • Circulating IgE binds to two isoforms of IgE receptors: high-affinity IgE receptors (Fc ⁇ RI) present on the surface of mast cells and basophils, and low affinity IgE receptors (Fc ⁇ RII or CD23) present on the surfaces of lymphocytes, eosinophils, platelets and macrophages. It is believed that an important factor governing the pathogenesis of allergic disorders is the cross-linkage of IgE receptors on mast cells, after encountering allergen and the consequent degranulation of mast cells.
  • Fc ⁇ RI high-affinity IgE receptors
  • Fc ⁇ RII or CD23 low affinity IgE receptors
  • the molecules released by mast cells include histamine, heparin, proteases and free radicals, which mediate a variety of biological effects including vasodilation, intestinal and/or bronchial smooth muscle contraction, mucous secretion and local proteolysis.
  • histamine histamine
  • heparin heparin
  • proteases free radicals
  • free radicals which mediate a variety of biological effects including vasodilation, intestinal and/or bronchial smooth muscle contraction, mucous secretion and local proteolysis.
  • the degranulation of IgE-dependent mast cells and the accumulation of eosinophils in the sites of inflammation are considered to result from the unbalanced overactivation of Th2 cells and consequently the Th2-mediated overproduction of IgE (Abbas et al., 1991, Nature 383:787-93; Vercelli, 2001, Curr Opin Allergy Clin Immunol 2001, 1:61-5).
  • the representative cytokines of Th2 cells such as IL-4, IL-5, IL-10 and IL-13, are known to play important roles in these reactions.
  • ThI -mediated cytokines such as IFN- ⁇ and IL- 12 were reported to negatively regulate the Th2 pathways.
  • IFN- ⁇ induces the isotype-switching to IgG2a in B cells
  • IL-12 converts the already established Th2 response to ThI dominance in certain situations
  • Various cellular transcription factors such as GAT A3 and T-bet, control the differentiation of ThI and Th2 cells and the production of cytokines in these cells (Lee et al., 2000, J Exp Med 192:105-15; Ting et al., 1996, Nature 384:474-8; Lighvani et al., 2001, Proc Natl Acad Sci USA 98:15137-42; Szabo et al., 2000, Cell 100:655-69).
  • GAT A3 and T-bet control the differentiation of ThI and Th2 cells and the production of cytokines in these cells (Lee et al., 2000, J Exp Med 192:105-15; Ting et al., 1996, Nature 384:474-8; Lighvani et al., 2001, Proc Natl Acad Sci USA 98:15137-42; Szabo et al., 2000, Cell 100:655-69).
  • Allergic diseases such as anaphylaxis, allergic rhinitis, asthma, atopic dermatitis, food allergies and urticaria, afflict up to 20% of the population in many countries and are increasing in prevalence (Wuthrich B., Int. Arch. Allergy Appl. Immunol. 90:3-10, 1989).
  • asthma is a significant disease of the lung which affects millions of people worldwide. Asthma is typically characterized by periodic airflow limitation and/or hyperresponsiveness to various stimuli which results in excessive airways narrowing. Other characteristics can include inflammation of airways, eosinophilia and airway fibrosis. Heightened airway responsiveness is thought to result from a complex inflammatory cascade involving several cell types, including T lymphocytes and eosinophils. In allergic asthma, Th2 cytokines predominate over ThI cytokines.
  • AD Atopic dermatitis
  • Th2 cells producing IL-4, IL-5, IL-IO, and IL-13 play a critical role in the initiating phase of the disease progression (Leung, 1997, Clin Exp Immunol 107(suppl. l):25-30).
  • IL-4 and IL- 13 act as major isotype inducers switching to IgE, and IL-5 induces the activation of eosinophils, which secretes various chemokines such as eotaxin.
  • IL-10 produced by monocytes/macrophages as well as Th2 cells augments the production of TARC, which is a Th2-specific chemokine and known to be overexpressed in AD lesions.
  • ThI -type cytokines such as IFN- ⁇
  • IFN- ⁇ Th2-mediated cytokines/chemokines and IgE
  • IFN- ⁇ and IL-12 the defective production of IFN- ⁇ and IL-12
  • ThI and Th2 types of reactions are reciprocally regulated in vivo
  • the modulation of Thl/Th2 has been thought to be a rational strategy for developing the therapeutics of allergic diseases (Kato et al., 1999, J Immunol 162:7470-9).
  • cytokines such as IL-12 and IFN- ⁇ or cytokine receptor antagonists to IL-4 and IL-5 have been tested for their ability to control the balance between ThI and Th2 responses (Hofstra et al., 1998, J Immunol 161:5054-60; Tomkinson et al., 2001, J Immunol 166:5792-800).
  • the direct administration of these agents often causes undesirable side effects.
  • Leukotrienes are also associated with a variety of diseases associated with inflammation, and particularly, with allergic inflammation. Leukotrienes are derived from arachidonic acid, the precursor of prostaglandins. There are two families of leukotrienes. The first group acts primarily in conditions in which inflammation is dependent on neutrophils, such as cystic fibrosis, inflammatory bowel disease, and psoriasis. The second group (cysteinyl-leukotrienes) is concerned primarily with eosinophil- and mast cell- induced bronchoconstriction in asthma.
  • Leukotrienes bind to highly selective receptors on bronchial smooth muscle and other airway tissue (O'Byrne et al., Annals of Internal Medicine 1997; 127:472-80).
  • Leukotrienes are also known to be important in the pathophysiology of allergic rhinitis, chronic urticaria and atopic dermatitis or eczema.
  • Leukotriene antagonists including both leukotriene synthesis inhibitors and cysteinyl-leukotriene receptor antagonists, are useful to specifically inhibit the production or actions of these inflammatory mediators.
  • Korea Patent Application No. 92-11752 disclosed an anti-inflammatory, anti-allergic and anti -rheumatic drug comprising biflavonoid such as 4'-O-methyl ochnaflavone isolated from Lonicera japonica, which shows efficacy in the treatment of various symptoms associated with allergy or inflammation.
  • Korea Patent Registration No. 100744 disclosed an anti-inflammatory, anti-allergic and anti-rheumatic drug comprising several biflavonoid compounds isolated from the leaves of Ginko biloba.
  • Several Oriental medicinal recipes comprising Siegesbeckia glabrescens have been reported to have IgE-reducing activity (Kim et al., Phytother. Res., 15:572-576, 2001).
  • many medicinal herbs have been found to be rich sources of histamine release inhibitors or anti-inflammatory compounds.
  • T cells T cells, the production of IgG2a, and the production of the associated ThI type cytokines (e.g., IFN- ⁇ , IL-6, IL-12, IL-I), is in contrast to the immune response associated with allergic inflammation discussed above.
  • ThI type cytokines e.g., IFN- ⁇ , IL-6, IL-12, IL-I
  • This type of immune response can have strong, systemic, anti-tumor and anti-viral properties. There is a continued need in the art for providing products with such properties.
  • One embodiment of the present invention relates to a method to regulate an immune response in a mammal.
  • the method includes administering a hardy kiwifruit preparation to the mammal in an amount sufficient to regulate an immune response in the mammal, wherein the hardy kiwifruit preparation is selected from: fresh fruit, crushed fruit, boiled fruit, cooked fruit, pressed fruit, condensed fruit, dried fruit, and a hardy kiwifruit juice concentrate.
  • the hardy kiwifruit preparation has not been extracted.
  • the hardy kiwifruit preparation is produced by a method that includes a step of drying the fruit.
  • Another embodiment of the present invention relates to a method to regulate an immune response in a mammal, including administering a hardy kiwifruit preparation to the mammal in an amount sufficient to regulate an immune response in the mammal, wherein the hardy kiwifruit preparation is selected from the group consisting of: a leaf extract or concentrate, and a bark extract or concentrate.
  • Yet another embodiment of the present invention relates to a method to regulate an immune response in a mammal.
  • the method includes administering to the mammal in an amount sufficient to regulate an immune response in the mammal: (a) a hardy kiwifruit preparation; and (b) a component selected from: probiotics; bacterial cell walls and fragments; whey protein; alanine; fatty acids; fatty acid esters; monoglycerides; diglycerides; triglycerides; inositol; turmeric; curcumin; methylsulfonylmethane (MSM); ginseng; ginger; and proanthocyanidin.
  • the hardy kiwifruit preparation can include, but is not limited to: a fruit extract or concentrate, a leaf extract or concentrate, a stem extract or concentrate, a bark extract or concentrate, a root extract or concentrate, fresh fruit, crushed fruit, boiled fruit, cooked fruit, pressed fruit, condensed fruit, dried fruit, a hardy kiwifruit juice concentrate, a preparation produced by extraction of fruit water having a temperature from O 0 C to about 80°C; a preparation produced by direct extraction of a water soluble concentrate of hardy kiwifruit with ethyl acetate, a preparation produced by extraction of hardy kiwifruit in distilled water, and a preparation produced by sequential extraction of hardy kiwifruit in water, chloroform and ethyl acetate.
  • the hardy kiwifruit preparation is produced by a method that includes a step of drying the fruit.
  • the hardy kiwifruit preparation is produced by extraction of fruit in water having a temperature of from about O 0 C to about 25°C.
  • the hardy kiwifruit preparation is produced by extraction of fruit in room temperature water.
  • the hardy kiwifruit preparation is produced by direct extraction of a water soluble concentrate of hardy kiwifruit with ethyl acetate.
  • Another embodiment of the present invention relates to a method to regulate an immune response in a mammal, comprising administering to the mammal in an amount sufficient to regulate an immune response in the mammal: (a) a hardy kiwifruit preparation; and (b) a component selected from: steroids, antihistamines, antibodies, antibiotics, cyclosporins, antimycotics, respiratory function controllers, analgesics, ⁇ -agonists, leukotriene modifiers, cytokine or cytokine receptor antagonists, phosphodiesterase inhibitors, sodium cromoglycate, nedocrimil, caffeine, theophylline, carbobenzoxy beta- alanyl taurine, and inhibitors of T cell function.
  • the hardy kiwifruit preparation can include, but is not limited to: a fruit extract or concentrate, a leaf extract or concentrate, a stem extract or concentrate, a bark extract or concentrate, a root extract or concentrate, fresh fruit, crushed fruit, boiled fruit, cooked fruit, pressed fruit, condensed fruit, dried fruit, a hardy kiwifruit juice concentrate, a preparation produced by extraction of fruit water having a temperature from 0°C to about 80° C; a preparation produced by direct extraction of a water soluble concentrate of hardy kiwifruit with ethyl acetate, a preparation produced by extraction of hardy kiwifruit in distilled water, and a preparation produced by sequential extraction of hardy kiwifruit in water, chloroform and ethyl acetate.
  • the hardy kiwifruit preparation is produced by a method that includes a step of drying the fruit.
  • the hardy kiwifruit preparation is produced by extraction of fruit in water having a temperature of from about 0°C to about 25°C.
  • the hardy kiwifruit preparation is produced by extraction of fruit in room temperature water.
  • the hardy kiwifruit preparation is produced by direct extraction of a water soluble concentrate of hardy kiwifruit with ethyl acetate.
  • the kiwifruit preparation can be provided in an amount sufficient to regulate a Th2 and a ThI immune response in the mammal.
  • the kiwifruit preparation is provided in an amount sufficient to regulate the amount of an antibody isotype produced by the mammal selected from the group consisting of IgE, IgG2a, and IgGl.
  • the kiwifruit preparation is provided in an amount sufficient to decrease the production and/or levels of at least one Th2 cytokine in the mammal or to increase the level of at least one ThI cytokine in the mammal, hi yet another aspect, the kiwifruit preparation is provided in an amount sufficient to decrease the level of or production of at least one leukotriene in the mammal. In another aspect, the kiwifruit preparation is provided in an amount sufficient to decrease the level of expression of a transcription factor selected from the group consisting of: GAT A-3, T-bet and NFATc2 in the mammal.
  • the mammal has or is at risk of developing a condition in which enhancement of a ThI response and/or suppression of a Th2 response is desirable.
  • the mammal can have or be at risk of developing an allergic disease or non-allergic inflammatory disease.
  • an allergic disease can be a disease that is regulated by leukotrienes.
  • allergic diseases include, but are not limited to, asthma and atopic dermatitis.
  • the mammal can have or be at risk of developing a viral infection or a cancer.
  • the hardy kiwifruit can include, but is not limited to: Actinidia arguta, Actinidia kolomikta and Actinidia polygama, with Actinidia arguta being one preferred embodiment.
  • the hardy kiwifruit preparation can be provided in a composition in an amount of between about 0.01% and about 95% by weight based on the total weight of the composition.
  • the step of administering comprises administering the hardy kiwifruit preparation with a carrier, adjuvant, or diluent to the mammal
  • the step of administering comprises providing the hardy kiwifruit preparation to the mammal as a tablet, a powder, an effervescent tablet, an effervescent powder, a capsule, a liquid, a suspension, a granule or a syrup.
  • the step of administering comprises providing the hardy kiwifruit preparation to the mammal in a health food.
  • Health foods include, but are not limited to: fine bakery wares, bread, rolls, breakfast cereals, processed cheese, unprocessed cheese, condiments, dairy products, puddings, gelatin desserts, carbonated drinks, teas, powdered beverage mixes, processed fish products, fruit-based drinks, vegetable-based drinks, chewing gum, hard confectionery, frozen dairy products, processed meat products, nut-based spreads, pasta, processed poultry products, gravies and sauces, potato chips, vegetable chips, crisps, chocolate, cookies, candy, licorice, ice creams, dehydrated foods, cut food products, processed food products, spices, alcoholic beverages, noodles, fermented foods, soups, soup mixes, soya based products, vegetable oil-based spreads, and vegetable-based drinks.
  • the step of administering comprises applying a cosmetic composition comprising the hardy kiwifruit preparation to the mammal.
  • Cosmetic compositions can be provided in a form including, but not limited to: lotion, cream, essence, toner, emulsion, pack, soap, shampoo, rinse, cleanser, body washing solution, washing solution or treatment.
  • the step of administering comprises providing the hardy kiwifruit preparation to the mammal in a food additive.
  • the method can further include administering to the mammal an agent selected from: fatty acids; polyketides; organic acids; small organic compounds; aromatic amino acids; phenylpropanoids; terpenoids; steroids; alkaloids; corrins; porphyrins; linear peptides; cyclic peptides; depsipeptides; amino acids derivatives; nucleosides; nucleotides; carbohydrates; proteins; cells; cell fragments; herbal preparations; spices; minerals; sterilizers; seasonings; vitamins; and electrolytes.
  • an agent selected from: fatty acids; polyketides; organic acids; small organic compounds; aromatic amino acids; phenylpropanoids; terpenoids; steroids; alkaloids; corrins; porphyrins; linear peptides; cyclic peptides; depsipeptides; amino acids derivatives; nucleosides; nucleotides; carbohydrates; proteins; cells; cell fragments; herbal preparations; spices; minerals; sterilizers; season
  • the method can further include administering to the mammal an agent selected from: probiotics; bacterial cell walls and fragments; whey protein; taurine; alanine; fatty acids; fatty acid esters; monoglycerides; diglycerides; triglycerides; inositol; turmeric; curcumin; rosemary; rosemarinic acid; methylsulfonylmethane (MSM); ginseng; ginger; proanthocyanidin; and ⁇ -carotene.
  • an agent selected from: probiotics; bacterial cell walls and fragments; whey protein; taurine; alanine; fatty acids; fatty acid esters; monoglycerides; diglycerides; triglycerides; inositol; turmeric; curcumin; rosemary; rosemarinic acid; methylsulfonylmethane (MSM); ginseng; ginger; proanthocyanidin; and ⁇ -carotene.
  • MSM methylsulfon
  • Fatty acids include, but are not limited to: conjugated linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, ⁇ -linolenic acid, ⁇ -linolenic acid, dihomo- ⁇ - linolenic acid, and stearidonic acid.
  • the method further includes administering to the mammal a different Actinidia species preparation.
  • the different Actinidia species can include, but is not limited to: A. chenensis, A. deliciosa, A. arguta, A. polygama, and A. kolomikta
  • Yet another embodiment of the present invention relates to a composition for regulating an immune response in a mammal.
  • the composition includes a hardy kiwifruit preparation and at least one additional active compound for regulating an immune response in a mammal.
  • the additional active compound is for treating or preventing allergic disease in a mammal.
  • the additional active compound is selected from: steroids, antihistamines, antibodies, antibiotics, cyclosporins, antimycotics, respiratory function controllers, analgesics, ⁇ -agonists, leukotriene modifiers, cytokine or cytokine receptor antagonists, phosphodiesterase inhibitors, sodium cromoglycate, nedocrimil, caffeine, theophylline, carbobenzoxy beta-alanyl taurine, and inhibitors of T cell function.
  • the additional active compound is selected from the group consisting of: probiotics; bacterial cell walls and fragments; whey protein; alanine; fatty acids; fatty acid esters; monoglycerides; diglycerides; triglycerides; inositol; turmeric; curcumin; methylsulfonylmethane (MSM); ginseng; ginger; and proanthocyanidin.
  • Fatty acids include, but are not limited to: conjugated linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, ⁇ -linolenic acid, ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid, and stearidonic acid.
  • a composition can include, but is not limited to, a pharmaceutical composition, a health food, a food additive, or a cosmetic.
  • the hardy kiwifruit can include, but is not limited to: Actinidia arguta, Actinidia kolomikta and Actinidia polygama.
  • the hardy kiwifruit preparation is an extract or concentrate prepared from a part of the hardy kiwifruit selected from: the fruit, the leaf, the stem, the bark, the root, and any combination thereof.
  • the hardy kiwifruit is selected from the group consisting of: fresh fruit, crushed fruit, boiled fruit, cooked fruit, pressed fruit, and condensed fruit.
  • the hardy kiwifruit is dried fruit, hi yet another aspect, the hardy kiwifruit preparation is produced by a method that includes a step of drying the fruit.
  • the hardy kiwifruit preparation is a hardy kiwifruit juice concentrate.
  • the hardy kiwifruit preparation is produced by extraction of fruit in room temperature water.
  • the hardy kiwifruit preparation is produced by direct extraction of a water soluble concentrate of hardy kiwifruit with ethyl acetate.
  • the extract is prepared by extraction of hardy kiwifruit in distilled water.
  • the extract is an ethyl acetate extract of the hardy kiwifruit.
  • Another embodiment of the invention relates to the use of hardy kiwifruit or a preparation thereof and an agent selected from: a steroid, an antihistamine, an antibody, an antibiotic, a cyclosporin, an antimycotic, a respiratory function controller, an analgesic, a ⁇ - agonists, a leukotriene modifier, a cytokine antagonist, a cytokine receptor antagonist, a phosphodiesterase inhibitor, sodium cromoglycate, nedocrimil, caffeine, theophylline, carbobenzoxy beta-alanyl taurine, and an inhibitor of T cell function, in the preparation of a composition for the treatment of a disease or condition that is associated with dysregulation of immune function.
  • an agent selected from: a steroid, an antihistamine, an antibody, an antibiotic, a cyclosporin, an antimycotic, a respiratory function controller, an analgesic, a ⁇ - agonists, a leukotriene modifier,
  • Another embodiment of the present invention relates to the use of hardy kiwifruit or a preparation thereof and an agent selected from: probiotics; bacterial cell walls and fragments; whey protein; alanine; fatty acids; fatty acid esters; monoglycerides; diglycerides; triglycerides; inositol; turmeric; curcumin; methylsulfonylmethane (MSM); ginseng; ginger; and proanthocyanidin.
  • an agent selected from: probiotics; bacterial cell walls and fragments; whey protein; alanine; fatty acids; fatty acid esters; monoglycerides; diglycerides; triglycerides; inositol; turmeric; curcumin; methylsulfonylmethane (MSM); ginseng; ginger; and proanthocyanidin.
  • Fatty acids include, but are not limited to: conjugated linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, ⁇ -linolenic acid, ⁇ - linolenic acid, dihomo- ⁇ -linolenic acid, and stearidonic acid.
  • the composition can be used for the treatment of a disease or condition that is associated with leukotriene production or activity.
  • the disease or condition can include, but is not limited to: atopic dermatitis, asthma, food allergy, allergic rhinitis, and chronic urticaria.
  • the hardy kiwifruit preparation can include, but is not limited to: a fruit extract or concentrate, a leaf extract or concentrate, a stem extract or concentrate, a bark extract or concentrate, a root extract or concentrate, fresh fruit, crushed fruit, boiled fruit, cooked fruit, pressed fruit, condensed fruit, dried fruit, a hardy kiwifruit juice concentrate, a preparation produced by extraction of fruit in room temperature water; a preparation produced by direct extraction of a water soluble concentrate of hardy kiwifruit with ethyl acetate, a preparation produced by extraction of hardy kiwifruit in distilled water, and a preparation produced by sequential extraction in water, chloroform and ethyl acetate.
  • Yet another embodiment of the present invention relates to a method to regulate an immune response in a mammal.
  • the method includes administering a common kiwifruit preparation to the mammal in an amount sufficient to regulate an immune response in the mammal, wherein the common kiwifruit preparation is selected from: a fruit extract or concentrate, a leaf extract or concentrate, a stem extract or concentrate, a bark extract or concentrate, a root extract or concentrate, fresh fruit, crushed fruit, boiled fruit, cooked fruit, pressed fruit, condensed fruit, dried fruit, a common kiwifruit juice concentrate, a preparation produced by extraction of fruit in room temperature water; a preparation produced by direct extraction of a water soluble concentrate of common kiwifruit with ethyl acetate, a preparation produced by extraction of common kiwifruit in distilled water, and a preparation produced by sequential extraction in water, chloroform and ethyl acetate.
  • Fig. 1 shows the inhibitory activity of various preparations from A. arguta on the production of IgE in U266B1 cells. Results were calculated as the percentage of IgE level produced from U266B1 cells treated with LPS only, from three-independent experiments.
  • Fig. 2 shows the dose-dependent effects of PG102T and PG102E on IL-4 production in OVA-stimulated splenocytes. Specific activity of PG102T and PG102E was determined from the IC 50 value.
  • Figs. 3A-3C illustrate the effects of PG102T and PGl 02E on the number of IL-4 or IFN- ⁇ -producing T cells (Fig. 3A) and IgE-producing B cells (Fig. 3B) and IgE biosynthesis within B cells (Fig. 3C). Data are the mean of percentages of each population from three independent experiments. *, P ⁇ 0.05 vs DW-treated mice.
  • Figs. 4A-4B show the effects of PG102T and PGl 02E on the expression of GATA3, T-bet and NFATc2, by Western blot (Fig. 4A) and quantitative real-time PCR analysis (Fig. 4B). Results are expressed as mean ⁇ SEM from three independent experiments. *, P ⁇ 0.05 and **, PO.01 VS. DW-treated mice, ⁇ -actin and GAPDH were used as a loading control.
  • Figs. 5A-5B show the effects of PGl 02T and PGl 02E on the development of dermatitis in NC mice using dermatitis index (Fig. 5A) and scratching incidence (Fig. 5B). Values are expressed as means ⁇ SEM of 5-6 animals. *, PO.05 ; **, PO.01, vs. DW- treated mice. Figs. 6A-6C show the effects of PG102T and PG102E on the plasma levels of IgE
  • Fig. 6A IgGl (Fig. 6B) and IgG2a (Fig. 6C) in NC mice. Values are expressed as mean ⁇ SEM of 5 animals. *, PO.05 ; **, PO.01, vs. DW-treated mice.
  • Figs. 7A-7B show the effects of PG102T and PG102E on the number of total leukocytes and eosinophils (Fig. 7A) and the production of eotaxin and TARC (Fig. 7B) in peripheral blood. Values are expressed as mean ⁇ SEM of 5 animals. *, PO.05 ; **, PO.01, vs. DW-treated mice.
  • Figs. 8A-8B show the effects of PG102T and PG102E on the skin lesions in NC mice from back skin (Fig. 8A) and face skin (Fig. 8B). *, PO.05 ; **, PO.01, vs. DW- treated mice.
  • Figs. 9A-9B show the effects of PG102T and PG102E on the expression of IL-4, IL-
  • Fig. 10 is a schematic of the process used in order to produce larger amounts of PGl 02T; this frozen or otherwise dried kiwifruit concentrate is also referred to as FDOOl (FG refers to food grade carrier).
  • Fig. 11 shows the effect of three doses (0.25, 1.0, and 10 mg/mL) of FDOOl
  • Cytokine levels were measure by ELISA. Each point represents the average of data from splenocytes of ten individual mice.
  • Fig. 12 shows the effect of three doses (0.25, 1.0, and 10 mg/mL) of an ethyl acetate
  • IL-4, IL-5, IL-10, IL-13, and IFN- ⁇ by OVA-stimulated mouse splenocytes following 3 days exposure in vitro. Cytokine levels were measure by ELISA. Each point represents the average of data from splenocytes of ten individual mice.
  • Fig. 13 shows the effect of three doses (0.25, 1.0, and 10 mg/mL) of an A. arguta fruit juice concentrate on the relative degree of production of the cytokines IL-4, IL-5, IL- 10, IL-13, and IFN- ⁇ by OVA-stimulated mouse splenocytes following 3 days exposure in vitro. Cytokine levels were measure by ELISA. Each point represents the average of data from splenocytes often individual mice.
  • Figs. 14A and 14B indicate the activity of three doses of known immunosuppressant compounds on the relative degree of production of IL-13, and IFN- ⁇ by OVA-stimulated mouse splenocytes following 3 days exposure in vitro.
  • Cyclosporin was tested at 0.0083, 0.083, and 4.15 ⁇ M and dexamethasone was tested at 0.01, 0.1, and 1 ⁇ M (Fig. 14A) with each point representing the average of data from splenocytes of ten individual mice.
  • Quercetin was tested at 1.0, 10, and 25 ⁇ M with each point representing the average of data from splenocytes of two individual mice (Fig. 14B). Cytokine levels were measure by ELISA.
  • Figs. 15A and 15B show the effect of three doses (1.0, 3.0, and 10 mg/mL) of FDOOl (PG102T), an ethyl acetate (EtOAc) extract of FDOOl, and the aqueous remainder from this process on the relative degree of production of IL-13 (Fig. 15A) and IFN- ⁇ (Fig.
  • Figs. 16A and 16B show the effect of three doses (1.0, 3.0, and 10 mg/mL) of FDOOl (PG102T) and a powdered form of FDOOl (created for use in capsules) on the relative degree of production of IL- 13 (Fig. 16A) and IFN- ⁇ (Fig. 16B) by OVA-stimulated mouse splenocytes following 3 days exposure in vitro. Cytokine levels were measure by ELISA. Each point represents the average of data from splenocytes of eight individual mice.
  • Figs. 17A and 17B show the effect of alternative preparations of A. arguta on the relative degree of production of IL- 13 (Fig. 17A) and IFN- ⁇ (Fig. 17B) by OVA-stimulated mouse splenocytes following 3 days exposure in vitro.
  • Three doses 1.0, 3.0, and 10 mg/mL) each of FDOOl (PG102T), a fruit juice concentrate, an. extract prepared by boiling of the fresh fruit in water, and a room temperature water extract of the fruit were tested.
  • Cytokine levels were measure by ELISA. Each point represents the average of data from splenocytes of eight individual mice.
  • Figs. 18A and 18B show the effect of preparations of alternative plant parts of A. arguta on the relative degree of production of IL- 13 (Fig. 18A) and IFN- ⁇ (Fig. 18B) by OVA-stimulated mouse splenocytes following 3 days exposure in vitro.
  • Three doses 1.0, 3.0, and 10 mg/mL
  • individual extracts prepared by boiling of the bark, root, or stem in H 2 O, and FDOOl were tested.
  • Cytokine levels were measured by ELISA. Each point represents the average of data from splenocytes of eight individual mice.
  • Figs. 19A-19C highlight the distributional shift that occurred between days 1 and 14 of a human clinical trial in which subjects responded positively to treatment for atopic dermatitis (AD) as measured by the Physician's Global Assessment (PGA) for AD (scoring criteria shown in Fig. 19C).
  • Subjects were administered placebo or 600 mg of FDOOl (PG102T) daily (Figs. 19A and 19B, respectively) and were using topical steroid treatment concomitantly.
  • PG102 the hardy kiwifruit, and particularly certain extracts/concentrates derived therefrom, generally referred to herein as PG102, increase serum levels of ThI cytokines and IgG2a, reduce serum levels of Th2 cytokines and IgE, inhibit histamine release from mast cells, and suppress allergic inflammatory reactions, including in an allergen-sensitized murine model of allergic inflammation and airway hyperresponsiveness, as well as in a rat paw edema assay (see U.S. Patent Publication No. 2004/0037909, supra). PG102 is orally active.
  • Examples 1 and 2 below exhibit inhibitory activity on the production of IgE as well as the ability to regulate selective ThI and Th2 cytokines. This regulatory activity is most likely achieved by the regulation of cellular transcription factors, GAT A-3, T-bet and NFATc2.
  • the present inventors' data indicates the great potential of PG102T and PG102E as natural immune modulators of the ThI and Th2 pathways, and ultimately as anti-allergy agents.
  • the present inventors also describe herein other preparations of hardy kiwifruit, including preparations of whole fruit, stem, root, bark, new extracts, concentrates, juices, dried preparations and non-extracted preparations, and demonstrate that such hardy kiwifruit preparations have similar properties with regard to their ability to regulate ThI and
  • Th2 cytokines are believed to therefore have similar anti-allergy properties.
  • compositions of the present invention can be used to treat leukotriene-mediated diseases, including, but not limited to, atopic dermatitis, asthma, food allergy, allergic rhinitis, and chronic urticaria.
  • the present inventors also demonstrate herein that extracts or other preparations of parts of the hardy kiwifruit plant other than the fruit itself have equivalent or superior immune regulatory activity as compared to the water soluble or ethyl acetate extracts of the fruit.
  • the present inventors have demonstrated the efficacy of extracts of the stem, root and bark of the hardy kiwifruit plant in suppressing cytokine production by antigen-stimulated splenocytes from allergen-sensitized mice.
  • the present inventors have also made the surprising discovery that preparations of hardy kiwifruit as described herein may serve as an effective adjunct to other therapies for various atopic conditions, including, but not limited to, steroid-based therapy.
  • preparations of hardy kiwifruit as described herein may serve as an effective adjunct to other therapies for various atopic conditions, including, but not limited to, steroid-based therapy.
  • the present inventors have discovered that administration of a powdered form of the water soluble extract of A. arguta as described herein to adult human patients suffering from atopic dermatitis of moderate severity, significantly reduced the physician's global rating of clinical signs.
  • the present invention in one embodiment, relates to the use of the hardy kiwifruit preparations described herein in combination with (or as an adjunct to) other therapeutic agents to treat atopic disease or other diseases associated with immune dysregulation.
  • compositions of the present invention can be used to enhance the efficacy of other therapeutic and nutritional therapies, particularly in patients with atopic conditions.
  • This embodiment of the invention is discussed in detail below.
  • the present inventors report herein that surprisingly, the process of drying hardy kiwifruit is a previously unappreciated, but important element to enhancing the bioactivity of the hardy kiwifruit.
  • the present inventors show herein that the dried hardy kiwifruit, extracted solely with room temperature water, exhibits similar bioactivity to that previously ascribed to hot water extracts or organic solvent extracts of hardy kiwifruit.
  • Dried hardy kiwifruit extracted in cool or cold water is also encompassed, thus extending extraction to any temperature between 0°C and 80°C.
  • the present inventors also set forth herein that dried hardy kiwifruit that has not been extracted (e.g., dried slices of hardy kiwifruit) can have the bioactivity that was previously ascribed to extracts of hardy kiwifruit. Therefore, the present invention relates to any form of dried hardy kiwifruit, including extracts produced from previously dried hardy kiwifruit, as an agent or for the preparation of a composition for regulating the immune response in a mammal. More specific uses for this agent or composition are described below.
  • the invention further relates to the use of any members of the family, Actinidiaceae, and particularly any members of the genus Actinidia, including, but not limited to, the common kiwi known as A. chinensis or A. deliciosa to provide compositions of kiwifruit with immune regulatory activity.
  • the present inventors believe that the process of drying other members of Actinidia can provide a composition of dried Actinidia having at least some of the biological activities that have been recognized for the hardy kiwifruit described herein.
  • the present inventors believe that other preparations of common kiwifruit (e.g., A. chinensis or A.
  • deliciosa including, but not limited to, preparations of any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract, concentrate, or fraction thereof, can have at least some of the biological properties that have been recognized for the hardy kiwifruit described herein.
  • A. arguta, A. polygama, and A. kolomikta belonging to Actinidiaceae are naturally distributed in Siberia, the northern area of China, North and South Korea. More than 30 species belonging to Actinidiaceae has been reported. Among those, the fruit of A. chinensis or A. deliciosa have been named "kiwi" and are popular edible fruits.
  • A. arguta and other fruit of the same genus e.g., A. polygama, and A.
  • reference to "hardy kiwifruit” refers to any of A. arguta, A. polygama, and A. kolomikta, or another species of the Actinidia genus related thereto that has the bioactive properties of A. arguta, A. polygama, and A. kolomikta as described herein, particularly with regard to the anti-allergy properties and the immune response/cytokine/leukotriene-regulatory properties demonstrated herein (e.g., see Examples).
  • any one or more parts of the hardy kiwifruit or other species of Actinidia including, but not limited to, the fruit (also referred to as the "berries” or the “kiwiberries"), the leaf, the stem, the bark and the root thereof.
  • an extract refers to a concentrated preparation of a substance ⁇ e.g., hardy kiwifruit) which is typically obtained by removing the active or desired constituents therefrom with a suitable solvent, and then evaporating some, all or nearly all the solvent and adjusting the residual mass or powder to a prescribed standard.
  • concentration refers to a form of substance which has had the majority of its base component, or solvent, removed. Accordingly, it will be apparent that the term “extract” as disclosed herein may be, in some embodiments, used interchangeably with the term “concentrate”.
  • Reference herein to a crude extract refers to, in one embodiment, an extract of hardy kiwifruit that is obtained by extracting a preparation of a hardy kiwifruit with water, lower alcohols (e.g., methanol, ethanol and the like), or the mixtures thereof, and preferably distilled water or 50-90% ethanol, and more preferably 70% ethanol.
  • a non- polar solvent soluble extract therefrom can be obtained by further extracting the soluble extract with a non-polar solvent such as hexane, ethyl acetate or dichloromethane solvent.
  • bioassays for tracking, evaluating or confirming the preferred biological activities of a hardy kiwifruit composition according to the present invention are set forth in the Examples herein, and include both in vitro and in vivo assays.
  • P102T refers generally to a total water-soluble extract (which may also be referred to herein as a concentrate) from a hardy kiwifruit described herein (e.g., A. argutd), prepared essentially as described in U.S. Patent Publication No. 2004/0037909, supra (e.g., see Example 1 of that publication), or as described in Example 1 herein, hi a preferred embodiment, the total water-soluble extract is prepared from A. arguta, although it will be apparent to those of skill in the art that equivalent total water-soluble extracts can be prepared from other hardy kiwifruit, including, but not limited to, A. polygama and A. kolomikta.
  • a different ethyl acetate extract is produced by directly extracting FDOOl (or PG 102T) with ethyl acetate ⁇ i.e., there is no chloroform extraction prior to the ethyl acetate fraction).
  • an ethyl acetate extract is prepared from A. arguta, although it will be apparent to those of skill in the art that equivalent extracts can be prepared from other hardy kiwifruit, including, but not limited to, A. polygama and A. kolomikta.
  • extracts or concentrates or other preparations of the invention can be produced from any species of Actinidia. Accordingly, it is an object of the present invention to provide a composition, including a pharmaceutical composition, a cosmetic composition, or a composition useful as or with a health food product, health food or beverage, or food additive (including human and animal (including domestic pet) food additives), that comprises such extracts of hardy kiwifruit or other species of Actinidia, if desired.
  • compositions are intended for use in any method of the invention, including to selectively regulate ThI and Th2 immune responses in a patient (i.e., in a mammal), such as the method for preventing or treating allergic and non-allergic inflammatory disease or providing an anti-viral or anti-cancer pharmaceutical or nutraceutical by administration of such compositions.
  • Other additives and components of the compositions, as well as the dosing and administration strategies described herein apply to this object of the invention as well.
  • any composition described herein in addition to the extracts described herein, including the extracts/concentrates described specifically above, included in the invention is the use of whole fruits of hardy kiwifruit, or fruit preparations that are processed, but not extracted, including, but not limited to, fresh fruit, crushed fruit (dried or fresh), boiled fruit (dried or fresh), cooked fruit, dried fruit, pressed fruit, frozen fruit, and condensed fruit.
  • a composition including a pharmaceutical composition, a cosmetic composition, or a composition useful as or with a health food product, health food or beverage, or food additive, that comprises such whole fruits of hardy kiwifruit or fruit preparations that may be processed in some manner (e.g., dried, boiled, etc.), but that are not necessarily extracted. Therefore, in one embodiment, the present invention relates to preparations of hardy kiwifruit (and common kiwifruit) that has not been extracted.
  • compositions described herein are intended for use in any method of the invention, including to selectively regulate ThI and Th2 immune responses in a patient (i.e., in a mammal), such as the method for preventing or treating allergic and non-allergic inflammatory disease or providing an anti-viral or anti-cancer pharmaceutical or nutraceutical by administration of such compositions.
  • Other additives and components of the compositions, as well as the dosing and administration strategies described herein apply to this object of the invention as well.
  • the juice of a hardy kiwifruit in addition to the extracts described herein, included in the invention is the use of the juice of a hardy kiwifruit, produced from any part of the hardy kiwifruit and by any suitable process.
  • the juice can be used as produced directly from the fruit (i.e., not diluted or concentrated), the juice can be diluted, or it can be concentrated to form a fruit juice concentrate.
  • fresh kiwifruit can be run through a conventional juicer. The juicer may remove the skins from the fruit resulting in a mixture of seeds, pulp, and juice.
  • compositions are intended for use in any method of the invention, and particularly in the method for preventing or treating allergic and non-allergic inflammatory disease or providing an anti-viral or anti-cancer pharmaceutical or nutraceutical by administration of such compositions.
  • Other additives and components of the compositions, as well as the dosing and administration strategies described herein apply to this object of the invention as well.
  • the hardy kiwifruit can be extracted in any temperature water ranging from 0°C to 80°C, including room temperature and cooler (e.g., from about 0°C to about 25°C).
  • a “dried hardy kiwifruit” or a “dried kiwifruit” includes any form of a hardy kiwifruit or other kiwifruit (e.g., common kiwi) that has been dried by any process.
  • the term "kiwifruit” can be used herein to generically refer to any member of the genus Actinidia, and includes the members of the hardy kiwifruit as discussed above, as well as members of the common kiwi, also as discussed above.
  • a dried kiwifruit includes any dried part of the kiwifruit (fruit, leaf, stem, root, etc.), and includes dried whole fruits, dried sliced fruit, dried crushed fruit, dried diced fruit, and dried condensed fruit, as well as any extracts of kiwifruit, wherein the material that is extracted is first dried prior to extraction.
  • the extracts themselves need not be dried or processed further, although that is generally the most useful form for the extracts for formulation into compositions and for storage.
  • Preferred methods of further concentration of the extracts for further use include, but are not limited to, evaporation, distillation, ultrafiltration, reverse osmosis, precipitation, adsorption to and elution from a stationary phase, and extraction into alternative solvents.
  • a preferred dried kiwifruit for use in the present invention is a dried kiwifruit preparation that is not subsequently extracted.
  • compositions and methods described herein apply to the use of any hardy kiwifruit, including the use of any part of the fruit, stem, leaf, bark or root of the hardy kiwifruit, or any extract or concentrate or fraction thereof, any form of the whole fruit or processed but not extracted fruit, fruit juice, or any extract or concentrate or fraction thereof, and further including hardy kiwifruit plant parts (the plant parts including the fruit,- stem, leaf, bark, and/or root) and plant part preparations that are prepared using a process that includes a step of drying.
  • compositions including a pharmaceutical composition, a nutraceutical composition, a food additive, a health food (including a beverage or a food material), or a cosmetic composition, comprising, consisting essentially of, or consisting of, the hardy kiwifruit described herein (i.e., including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract, concentrate or fraction thereof).
  • the present invention provides a crude extract, a total water- soluble extract, or an ethyl acetate extract of the hardy kiwifruit.
  • the hardy kiwifruit preparation is an active ingredient for use to selectively regulate ThI and Th2 immune responses in a patient (i.e., in a mammal).
  • the composition has a biological activity selected from at least one of the following activities: (a) reduces the number of IgE-producing B cells in a patient; (b) reduces the amount of IgE produced in a patient (e.g., in the serum or plasma); (c) decreases production and/or levels of at least one Th2 cytokine (e.g., IL-4, IL-5, IL-IO); (d) increases the level of at least one ThI cytokine (e.g., IL- 12, IFN- ⁇ ); (e) decreases the level of expression of the transcription factor, GATA- 3; (f) increases the level of expression of the transcription factor T-bet; (g) increases the level of expression of the transcription factor NFATc2; (h) increases the number of IgG2a- producing B cells
  • the phrase "protected from a disease” refers to reducing the symptoms of the disease; reducing the occurrence of the disease, and/or reducing the severity of the disease.
  • Protecting a patient can refer to the ability of a composition of the present invention, when administered to a patient, to prevent a disease from occurring and/or to cure or to alleviate at least one, and preferably more than one, disease symptoms, signs or causes.
  • to protect a patient from a disease includes both preventing disease occurrence (prophylactic treatment) and treating a patient that has a disease (therapeutic treatment) to reduce the symptoms of the disease.
  • protecting a patient from a disease or enhancing another therapy is accomplished by regulating a given activity such that a beneficial effect is obtained.
  • disease refers to any deviation from the normal health of a mammal and includes a state when disease symptoms are present, as well as conditions in which a deviation (e.g., infection, gene mutation, genetic defect, etc.) has occurred, but symptoms are not yet manifested.
  • the biological activity or biological action of an agent described herein refers to any function(s) exhibited or performed by the agent that is ascribed to the naturally occurring form of the agent as measured or observed in vivo (i.e., in a natural physiological environment wherein the agent is used) or in vitro (i.e., under laboratory conditions).
  • Modifications of an agent such as by changing the processing or preparation of the agent or purification of the agent, may result in agents having the same biological activity as the naturally occurring agent, or in agents having decreased or increased biological activity as compared to the naturally occurring agent.
  • a composition including a pharmaceutical composition or a nutraceutical (nutritional) composition, comprising the hardy kiwifruit described herein (i.e., including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof), and in one embodiment, a crude extract, a total water-soluble extract, or an ethyl acetate extract of the hardy kiwifruit (which can collectively be referred to as active ingredients of the invention or as hardy kiwifruit preparations of the invention), as an active ingredient for regulating an immune response in the mammal, and more particularly for regulating a Th2 and/or a ThI immune response in a mammal, and even more particularly, for enhancing a ThI response in a mammal and/or suppressing a Th
  • Such active ingredients are useful for the treatment and/or prevention of a variety of conditions and diseases, including, but not limited to, allergic disease, non-allergic inflammatory disease, viral infection and cancer.
  • the composition can further comprise or be used in conjunction with additional therapeutic or nutraceutical agents for the prevention, treatment, and/or improvement of any of the above-described conditions or diseases.
  • nutritional applications include any applications of the invention directed to the provision of nutrients and nutritional agents to maintain, stabilize, enhance, strengthen, or improve the health of an individual or the organic process by which an organism assimilates and uses food and liquids for functioning, growth and maintenance, and which includes nutraceutical applications.
  • Therapeutic applications include any applications of the invention directed to prevention, treatment, management, healing, alleviation and/or cure of a disease or condition that is a deviation from the health of an individual.
  • Other applications of the invention include, for example, cosmetic applications. It is also an object of the present invention to provide a hardy kiwifruit described herein (i.e.
  • a hardy kiwifruit preparation including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof), and in one embodiment, a crude extract, a total water-soluble extract, or an ethyl acetate extract of the hardy kiwifruit, for the preparation of a therapeutic or nutraceutical agent for regulating an immune response in the mammal, and more particularly for regulating a Th2 and/or a ThI immune response in a mammal, and even more particularly, for enhancing a ThI response in a mammal and/or suppressing a Th2 response in a mammal.
  • Such active ingredients are useful for the treatment and/or prevention of a variety of conditions and diseases, including, but not limited to, allergic disease, non-allergic inflammatory disease, viral infection and cancer.
  • the agent can be used in conjunction with additional therapeutic or nutraceutical agents for the prevention, treatment, and/or improvement of any of the above-described conditions or diseases.
  • Such health food or health food additives are useful for the treatment and/or prevention of a variety of conditions and diseases,
  • an animal feed or feed additive comprising the hardy kiwifruit described herein (i.e., a hardy kiwifruit preparation, including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof), and in one embodiment, a crude extract, a total water- soluble extract, or an ethyl acetate extract of the hardy kiwifruit, as an essential component for regulating an immune response in the mammal, and more particularly for regulating a Th2 and/or a ThI immune response in a mammal, and even more particularly, for enhancing a ThI response in a mammal and/or suppressing a Th2 response in a mammal.
  • Such animal feeds or animal feed additives are useful for the treatment and/or prevention of a variety of conditions and diseases
  • It is still another object of the present invention to provide a topical or cosmetic composition comprising the hardy kiwifruit described herein (i.e. a hardy kiwifruit preparation, including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or traction thereof), and in one embodiment, a crude extract, a total water- soluble extract, or an ethyl acetate extract of the hardy kiwifruit, for regulating an immune response in the mammal, and more particularly for regulating a Th2 and/or a ThI immune response in a mammal, and even more particularly, for enhancing a ThI response in a mammal and/or suppressing a Th2 response in a mammal.
  • Such cosmetic compositions are useful for the treatment and/or prevention of a variety of conditions and diseases, including, but not limited
  • compositions, additives, or agents described herein may additionally comprise at least one conventional carrier, adjuvant or diluent.
  • the composition according to the present invention can include pharmaceutically acceptable carriers, adjuvants or diluents, e.g., lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starches, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinyl pyrrolidone, water, methylhydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate and mineral oil.
  • pharmaceutically acceptable carriers, adjuvants or diluents e.g., lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol,
  • the formulations may additionally include fillers, anti-agglutinating agents, lubricating agents, wetting agents, flavoring agents, emulsifiers, preservatives and the like.
  • the compositions of the invention may be formulated so as to provide quick, sustained or delayed release of the active ingredient after their administration to a patient.
  • the compositions of the present invention can be dissolved in oils, propylene glycol or other solvents that are commonly used to produce an injection. Suitable examples of the carriers include physiological saline, polyethylene glycol, ethanol, vegetable oils, isopropyl myristate, etc., but are not limited to these carriers.
  • the compounds of the present invention can be formulated in the form of ointments and creams.
  • compositions or formulations of the present invention may be prepared in any form, such as oral dosage form (effervescent tablet, effervescent powder, powder, tablet, capsule, soft capsule, aqueous medicine, syrup, elixirs pill, powder, sachet, granule), or topical preparation (cream, ointment, lotion, gel, balm, patch, paste, spray solution, aerosol and the like), or injectable preparation (solution, suspension, emulsion).
  • oral dosage form effervescent tablet, effervescent powder, powder, tablet, capsule, soft capsule, aqueous medicine, syrup, elixirs pill, powder, sachet, granule
  • topical preparation cream, ointment, lotion, gel, balm, patch, paste, spray solution, aerosol and the like
  • injectable preparation solution, suspension, emulsion
  • composition of the present invention in pharmaceutical dosage forms may be used alone or in appropriate association or combination with other pharmaceutically active compounds, including anti-inflammatory compounds, anti-allergy compounds, or any other compounds or compositions that can regulate an immune response or provide a benefit to a patient.
  • pharmaceutically active compounds including anti-inflammatory compounds, anti-allergy compounds, or any other compounds or compositions that can regulate an immune response or provide a benefit to a patient.
  • Compounds that are particularly desirable for use in the compositions and formulations of the present invention are described in detail herein.
  • composition of the present invention can also be provided as a health food that includes the hardy kiwifruit preparations of the invention (e.g., various foods, beverage, gum, vitamin complex, health improving food and the like).
  • the health food can be provided as a food item, powder, granule, tablet, chewing tablet, capsule or beverage etc.
  • Child or infant foods are also included in the compositions of the invention, such as modified milk powder, infant formulas, and modified infant or children's food.
  • Suitable food products into which a composition or agent of the invention can be introduced to produce a health food product include, but are not limited to, fine bakery wares, bread and rolls, breakfast cereals, processed and unprocessed cheese, condiments (ketchup, mayonnaise, etc.), dairy products (milk, yogurt), puddings and gelatin desserts, carbonated drinks, teas, powdered beverage mixes, processed fish products, fruit-based drinks (including fruit juices), vegetable-based drinks (including vegetable juices), chewing gum, hard confectionery, frozen dairy products, processed meat products, nut and nut-based spreads, pasta, processed poultry products, gravies and sauces, potato chips and other chips or crisps, chocolate and other confectionery (cookies, candy, licorice), ice creams, dehydrated foods, cut or processed food products (e.g., fruits, vegetables), spices, alcoholic beverages, noodles, fermented foods, soups and soup mixes, soya based products (milks, drinks, creams, whiteners), vegetable oil-based spreads, and
  • compositions described above, and particularly cosmetic formulations containing the above-identified compositions may be prepared in any form such as skin, lotion, cream, essence, toner, emulsion, pack, soap, shampoo, rinse, cleanser, body washing solution, washing solution, treatment, gel, balm, spray solution and the like.
  • compositions of the present invention can further include one or more of lactose, casein, dextrose, glucose, sucrose and sorbitol.
  • Any of the compositions, preparations, additives, or agents described herein may additionally comprise at least one active agent ⁇ i.e., active compound, active component).
  • the additional active agent can be a pharmacologically active agent and/or a nutritionally active agent.
  • Active agents typically contribute at least one additional desirable, nutritional, and/or therapeutic and/or pharmacological property to a composition, in addition to the kiwifruit preparation described herein. Active agents can be included in a composition, preparation, additive, or other formulation of the invention in any effective amount.
  • An effective amount is an amount sufficient to achieve the desired effect imparted by the agent, such as an effect on the health or nutrition of a subject (e.g., a therapeutic or nutritional effect), a taste effect, an aroma effect, a visual effect, etc.
  • an effect on the health or nutrition of a subject e.g., a therapeutic or nutritional effect
  • a taste effect e.g., a taste effect
  • an aroma effect e.g., a visual effect
  • One of skill in the art will be able to determine the appropriate amount of additional agents to add to a composition of the invention.
  • any of the compositions provided by or useful in the present invention can include one or more natural products as an active agent, including, but not limited to, fatty acids and polyketides; organic acids and miscellaneous small organic compounds; aromatic amino acids and phenylpropanoids; terpenoids and steroids; alkaloids; corrins and porphyrins; linear and cyclic peptides, depsipeptides, and other amino acids derivatives; nucleosides and nucleotides; carbohydrates; proteins, cells and cell fragments; herbal preparations and spices; minerals; sterilizers; seasonings; vitamins; electrolytes; and other natural agents.
  • natural products including, but not limited to, fatty acids and polyketides; organic acids and miscellaneous small organic compounds; aromatic amino acids and phenylpropanoids; terpenoids and steroids; alkaloids; corrins and porphyrins; linear and cyclic peptides, depsipeptides, and other amino acids derivatives; nucleosides and nu
  • compositions of the invention include synthetic flavoring agents, a coloring agent, a processing agent, an alginic acid or the salt thereof, an organic acid, a protective colloidal adhesive, a pH controlling agent, stabilizer, a preservative, a glycerin, an alcohol, a carbonation agent, or any other essential agent of a formulation (for nutritional or therapeutic use by any method of administration), a food, or a beverage.
  • Particularly preferred components (active agents) to combine with a hardy kiwifruit preparation of the invention or add to a composition containing such preparation include, but are not limited to: probiotics; bacterial cell walls and fragments; whey protein; taurine; alanine; fatty acids (e.g., conjugated linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, ⁇ -linolenic acid, ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid, stearidonic acid); mono-, di-, and triglycerides (composed of any combination of the fatty acids described above); inositol; turmeric; curcumin; rosemary; rosemarinic acid; methylsulfonylmethane (MSM); ginseng; ginger; proanthocyanidin; ⁇ -carotene; and any other preparation of a different species of kiwifruit than that used as the
  • Fatty acids and polyketides include, but are not limited to: saturated fatty acids (e.g., ⁇ -lipoic acid (R, S, or R,S); unsaturated fatty acids (e.g., conjugated linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, ⁇ -linolenic acid, ⁇ -linolenic acid, dihomo- ⁇ - linolenic acid, stearidonic acid); fatty acid esters; monoglycerides, diglycerides, and triglycerides (composed of any combination of the fatty acids described above); acetylenic fatty acids; branched-chain fatty acids; prostaglandins; thromboxanes; leukotrienes; aromatic polyketides; macrolides and polyethers; lipid extracts (e.g., marine oils, Echium oil, borage oil, olive oil); and lecithin.
  • Organic acids and miscellaneous small organic compounds include, but are not limited to, citric acid; fumaric acid; guaiacol; methylsulfonylmethane (MSM); and ascorbic acid.
  • Aromatic amino acids and phenylpropanoids include, but are not limited to, aromatic amino acids and benzoic acids (e.g., benzoic acid, gallic acid, gentisic acid, p- hydroxybenzoic acid, protocatechuic acid, vanillic acid, salicylic acid, syringic acid); cinnamic acids (e.g., hydroxytyrosol, curcumin, rosmarinic acid, ⁇ r-turmerone, caffeic acid, eugenol, chlorogenic acid, neochlorogenic acid, cinnamic acid, feralic acid, o-coumaric acid, /3-coumaric acid); lignans and lignin; phenylpropenes; coumarins; styrylpyrones; flavonoids (e.g., anthocyanidins, such as delphinidin; proanthocyanidins; catechins such as catechin, epicatechin, and thea
  • Terpenoids and steroids include, but are not limited to, monoterpenes (e.g., ⁇ - pinene, borneol, carvacvol, geraniol, thymol, 1,8-cineol, terpineol); iridoids (e.g., monotropein); ⁇ -ionone (e.g., thirteen carbon precursor to A vitamins); sesquiterpenes (e.g., caryophyllene, farnesol); diterpenes (e.g., A vitamins); sesterterpenes; triterpenes (e.g., ⁇ - amyrin, lupeol, ursolic acid); tetraterpenes; carotenoids (e.g., lycopene, ⁇ -carotene, lutein, astaxanthin, canthaxanthin); and steroids (e.g., D vitamins, ⁇ -sitosterol).
  • monoterpenes e.
  • Alkaloids include, but are not limited to, pyrrolidine alkaloids, tropane alkaloids, pyrrolizidine alkaloids, piperidine alkaloids, quinolizidine alkaloids, indolizidine alkaloids, pyridine alkaloids, phenylethylamines, tetrahydroisoquinoline alkaloids, galanthamines, indole alkaloids, ⁇ -carboline alkaloids, terpenoid indole alkaloids, quinoline alkaloids, pyrroloindole alkaloids, ergot alkaloids, quinazoline alkaloids, quinoline and acridine alkaloids, imizadole alkaloids, piperidine alkaloids, ephedrines, capsaisins, pyridine monoterpene alkaloids, aconitines, steroidal alkaloids, purine alkaloids (e.g., allantoin, caffeine, theophylline).
  • Corrins and porphyrins include, but are not limited to, B vitamins.
  • Linear and cyclic peptides, depsipeptides, and other amino acids derivatives include, but are not limited to, simple amino acids and their derivatives (e.g., L-acetyl carnitine, choline, taurine, alanine), linear peptides, cyclic peptides (e.g., cyclosporins), cyclic depsipeptides, ⁇ -lactams, cyanogenic glycosides, glucosinolates, cysteine sulphoxides.
  • Carbohydrates include, but are not limited to, monosaccharides (e.g., inositol), polysaccharides (e.g., fracto-oligosaccharides, such as inulin (any chain length); galacto- oligosaccharides; chitin and chitosan).
  • monosaccharides e.g., inositol
  • polysaccharides e.g., fracto-oligosaccharides, such as inulin (any chain length); galacto- oligosaccharides; chitin and chitosan.
  • proteins e.g., whey protein and superoxide dismutase
  • cells and cell fragments e.g., probiotics, meaning live, intact microorganisms such as, e.g., Lactobacillus spp., bacterial cells and cell wall fragments, fungal/yeast cells and cell wall fragments
  • herbal preparations and spices e.g., ginseng, huang, turmeric, rosemary, ginger
  • minerals e.g., K, Mg, Ca, Mn, Fe, Cu, Zn, B, Si, Se).
  • Metabolites and derivatives of any of these compounds are also encompassed by the present invention.
  • composition of the invention is administered as an adjunct therapy for a conventional therapy for a condition or disease.
  • a conventional therapy for a condition or disease for example, the present inventors have demonstrated that preparation of hardy kiwifruit according to the present invention improves the clinical outcome in patients with atopic dermatitis when used as an adjunct to topical steroid therapy. Therefore, compositions of the invention may include one or more therapeutic agents (e.g., medicines), which can also be referred to herein as active agents, used to treat a condition or disease that can be treated or ameliorated by regulation of immune responses.
  • therapeutic agents e.g., medicines
  • active agents used to treat a condition or disease that can be treated or ameliorated by regulation of immune responses.
  • Such therapeutic agents include, but are not limited to, steroids (including corticosteroids, and including oral, inhaled and injected), antihistamines (any type, including systemic, topical, inhaled, and including Hl and H2 blockers), antibodies (e.g., anti-IgE, anti-IL-10), antibiotics, cyclosporins, antimycotics, respiratory function controllers, analgesics, ⁇ -agonists (long or short acting), leukotriene modifiers (inhibitors or receptor antagonists), cytokine or cytokine receptor antagonists, phosphodiesterase inhibitors, sodium cromoglycate, nedocrimil, theophylline, caffeine, carbobenzoxy beta-alanyl taurine, inhibitors of T cell function and other anti-inflammatory agents.
  • steroids including corticosteroids, and including oral, inhaled and injected
  • antihistamines any type, including systemic, topical, inhaled, and including Hl and H2 blockers
  • antibodies
  • compositions may additionally comprise one or more than one organic acid (e.g., citric acid, fumaric acid, adipic acid, lactic acid, malic acid, ascorbic acid), phosphate (e.g., phosphate, sodium phosphate, potassium phosphate, acid pyrophosphate, polyphosphate), and/or natural anti-oxidants (e.g., polyphenol, catechin, ⁇ - tocopherol, rosemary extract, vitamin C, green tea extract, licorice root extract, chitosan, tannic acid, phytic acid, etc).
  • organic acid e.g., citric acid, fumaric acid, adipic acid, lactic acid, malic acid, ascorbic acid
  • phosphate e.g., phosphate, sodium phosphate, potassium phosphate, acid pyrophosphate, polyphosphate
  • natural anti-oxidants e.g., polyphenol, catechin, ⁇ - tocopherol, rosemary extract, vitamin C, green tea extract,
  • compositions of the present invention and particularly cosmetic compositions or compositions that are formulated for topical administration, including therapeutic compositions (but not limited to cosmetic or other topical compositions) can comprise additional additives including, but not limited to, water soluble vitamin, lipid soluble vitamin, peptide polymer, polysaccharide polymer, sphingolipid, glycosaminoglycans, B- glucan and sea-weed extract.
  • additional additives including, but not limited to, water soluble vitamin, lipid soluble vitamin, peptide polymer, polysaccharide polymer, sphingolipid, glycosaminoglycans, B- glucan and sea-weed extract.
  • Such compositions can be used as creams, lotions, massage packs or oils, and body washing solutions, soap, shampoos and the like.
  • Preferred water soluble vitamins are any one which can be mixed with cosmetics or other topical formulations, however, various vitamin such as vitamin Bl, B2, B6, pyridoxine, pyridoxine HCl, vitamin B 12, pantothenic acid, nicotinic acid, nicotinamide, folic acid, vitamin C, vitamin H etc, their salt thereof such as thiamin HCl salt, ascorbic acid Na salt etc or their derivatives thereof such as ascorbic acid-2-phosphonic acid Na salt, ascorbic acid-2-phosphonic acid Mg salt are preferable, and those can be obtained by conventional methods such as microbial conversion methods, purification methods from the microbial cultivates, enzymatic methods or chemical synthetic methods.
  • various vitamin such as vitamin Bl, B2, B6, pyridoxine, pyridoxine HCl, vitamin B 12, pantothenic acid, nicotinic acid, nicotinamide, folic acid, vitamin C, vitamin H etc, their salt thereof such as thia
  • Preferred lipid soluble vitamins are any one which can be mixed with cosmetics or other topical formulations, however, various vitamin such as vitamin A, D2, D3, E (dl- ⁇ - tocopherol, d- ⁇ -tocopherol, d- ⁇ -tocopherol) and their derivatives such as palmitic acid ascorbate, stearic acid ascorbate, dipalmitic acid ascorbate, acetic acid-dl- ⁇ -tocopherol, nicotinic acid dl- ⁇ -tocopherol vitamin E, dl-pantothenyl alcohol, d-pantothenyl alcohol, pantothenyl ethylether etc. containing the lipid soluble vitamin used in examples of present invention are preferable and those can be obtained by conventional methods such as microbial conversion methods, purification methods from the microbial cultivates, enzymatic methods or chemical synthetic methods.
  • Preferred peptide polymers are any one which can be mixed with cosmetics or other topical formulations; however, collagen, hydrolysable collagen, gelatin, elastin, hydrolysable gelatin, or keratin, etc., containing the peptide polymer used in examples of present invention are preferable.
  • Preferred polysaccharide polymers are any one which can be mixed with cosmetics or other topical formulations, however, hydroxy ethyl cellulose, xanthin gum, hyaluronic acid Na, chondroitin sulfate or their salt (Na salt etc) and the like are preferable.
  • chondroitin sulfate or the salt thereof etc. can be used by being purified from mammals or fish ordinarily.
  • Preferred sphingolipids are any one which can be mixed with cosmetics or other topical formulations, however, squalane, ceramide, pit-sphingosin, sphingo- lipopolysacchari.de and the like are preferable.
  • Sphingo-lipids can be obtained by purification from mammals, fish, shellfish, yeast, or plants etc. using conventional methods.
  • Preferred seaweed extracts are any one which can be mixed with cosmetics or other topical formulations, however, the extract of brown algae, red algae, green algae and the like, or the purified carrageenan, alginic acid, arginic acid, Na, K, or glycosaminoglycans isolated therefrom are preferable.
  • Algal extracts can be obtained by purification from seaweed using conventional methods.
  • the cosmetic and other topical compositions of the present invention may be combined with other ingredients or combined with a conventional cosmetic or topical composition, if necessary, together with above described hardy kiwifruit preparations.
  • Such other ingredients include, but are not limited to, oil ingredients, humectants, emollients, surface active agents, organic or inorganic dye, organic powder, ultraviolet ray absorbing agent, preservatives, antiseptics, antioxidants, plant extract, pH controller, alcohol, pigments, perfumes, refrigerants, antihidrotic, distilled water etc.
  • Preferable oil ingredients may comprise ester oil, hydrocarbon oil, silicone oil, fluoride oil, animal oil, plant oil and so on.
  • Preferred ester oils include, but are not limited to, glyceryl tri-2-ethyl hexanoic acid, cetyl 2-ethyl hexanoic acid, isopropyl myristic acid, butyl myristic acid, isopropyl palmitic acid, ethyl stearic acid, octyl palmitic acid, isocetyl isostearic acid, butyl stearic acid, ethyl linoleic acid, isopropyl linoleic acid, ethyl oleic acid, isocetyl myristic acid, isostearyl myristic acid, isostearyl palmitic acid, octyldodecyl myristic acid, isocetyl isostearic acid, diethyl sebasic acid, isopropyl adipic acid, isoalkyl neopetanoic acid, gly
  • Preferred silicone oils may comprise polymethylsilicone, methylphenylsilicone, methylcyclopolysiloxane, octamethylpolysiloxane, decamethylpolysiloxane, dodecamethylcyclosiloxane, dimethyl siloxane-methyl cetyl oxysiloxane copolymer, dimethyl siloxane-methyl stealloxysiloxane copolymer, alkyl modified silicone oil, amino modified silicone oil and the like.
  • Preferred fluoride oil can comprise perfluoropolyether and the like.
  • Preferred animal or plant oils can comprise avocado oil, almond oil, olive oil, sesame oil, rice husk oil, safflower oil, soybean oil, corn oil, rapeseed oil, canola oil, palm kernel oil, palm oil, sunflower oil, cotton seed oil, coconut palm oil, cucui nut oil, wheat embryo bud oil, rice embryo bud oil, shea butter, evening-primrose oil, macadamia nut oil, menhaden oil and other fish body oils, egg yolk oil, lanolin, hempseed oil, mink oil, orange roughy oil, jojoba oil, carnauba wax, liquid lanolin and the like.
  • Preferred bumectants can comprise water-soluble low molecular humectants, lipophilic low molecular humectants, water-soluble polymer and lipid soluble polymer.
  • preferable water soluble low molecular humectants can comprise cerin, glutamine, sorbitol, mannitol, pyrrolidone-carboxylic acid Na, glycerin, propylene glycol, 1,3-butylene glycol, ethylene glycol, polyethylene glycol (polymerization index. >2), polypropylene glycol (polymerization index >2), lactic acid, lactate salt and the like.
  • Preferred lipid soluble low molecular humectants can comprise cholesterol, cholesteryl ester and the like.
  • Preferred water-soluble polymers can comprise carboxy vinyl polymer, poly asparaginic acid salt, tragacanth, xanthin gum, HMC (hydroxy methyl celluose), HEC (hydroxy ethyl celluose), HPC (hydroxy propyl celluose), carboxymethylcellulose, water- soluble chitin, chitosan, dextrin and the like.
  • Preferred lipid soluble polymers can comprise polyvinylpyrrolidone-eicocene copolymer, polyvinylpyrrolidone-hexadecene copolymer, nitrocellulose, dextrin fatty acid ester, silicone polymer and the like.
  • Preferred emollients can comprise long chain acyl glutamic acid cholesteryl ester, cholesteryl hydroxy stearic acid, 12-hydroxy stearic acid, rogic acid, lanolin fatty acid cholesteryl ester and the like.
  • Preferred surface-active agents can comprise nonionic surfactants, anionic surfactants, cationic surfactants, amphivalent surfactants and the like.
  • preferred non-ionic surfactants can comprise self-emulsified monostearic acid glycerin, propylene glycol fatty acid ester, glycerin fatty acid ester, polyglycerin fatty acid ester, sorbitan fatty acid ester, polyoxyethylene (POE) sorbitan fatty acid ester, POE sorbitan fatty acid ester, POE glycerin fatty acid ester, POE alkyl ether, POE fatty acid ester, POE solid pimaja oil, POE pimaja oil, POE-POP copolymer, POE- POP alkyl ether, polyether modified silicone, lauric acid alkanol amide, alkyl amine oxide, hydrogen addition soybean phospholipid and the like.
  • POE polyoxyethylene
  • Preferred anionic surfactants can comprise fatty acid soap, ⁇ -acyl sulfonic acid salt, alkyl sulfonic acid salt, alkyl ally sulfonic acid, alkyl naphthalene sulfonic acid salt, alkyl sulfonic acid salt, POE alkylether sulfate salt, alkyl amide sulfate salt, alkyl phosphate salt, POE alkyl phosphate salt, alkylamide phospahate salt, alkyloylalkyl taurine salt, N-acyl- amino acid salt, POE alkyl ether carboxylic acid salt, alkyl sulfo succinic aid salt, alkyl sulfo-acetic acid salt, acylated hydrolysable collagen peptide salt, perfmoro alkyl phosphate ester and the like.
  • Preferred cationic surfactants can comprise alkyl trimethyl ammonium chloride, stearyl trimethyl ammonium chloride, stearyl trimethyl ammonium bromide, setostearyltrimethyl ammonium chloride, distearyl dimethyl ammonium chloride, stearyl dimethyl benzyl ammonium chloride, phenyl trimethyl ammonium bromide, benzalkonium chloride, diethylamino ethyl amide stearic acid, dimethylaminopropyl amide stearic acid, lanolin derivatives quaternary ammonium and the like.
  • Preferred ambivalent surfactants can comprise carboxy betaine type, amide betaine type, hydroxy sulfo betaine type, phosphpobetaine type, aminocarboxylic acid, imidazoline derivatives type, amide amine type and the like.
  • Preferred organic and inorganic dyes can comprise silicic acid, anhydrous silicic acid, magnesium silicic acid, talc, ceracyte, mica, kaolin, bengala, clay, bentonite, titan film mica, oxy chlorine bismuth, zirconium oxide, magnesium oxide, zinc oxide, titan oxide, aluminum oxide, calcium sulfate, barium sulfate, magnesium sulfate, calcium carbonate, magnesium carbonate, ferrous oxide, chromium oxide, chromium hydroxide, calamine, carbon black and combinations thereof as an inorganic dyes; polyamide, polyester, polypropylene, polystyrene, polyurethane, vinyl resin, urea resin, phenol resin, fluoride resin, silicone resin, acryl resin, melamine resin, epoxy resin, polycarbonate resin, divinyl benzene-styrene copolymer, silk powder, cellulose, CI pigment yellow, CI pigment orange as an organic dyes; and their complex etc
  • Preferred organic powders can comprise metal soaps such as calcium stearate; alkyl phosphonate metal salt such as sodium zinc cetylic acid, zinc laurylic acid, calcium laurylic acid; acylamino acid polyvalent metal salt such as calcium N-lauroyl- ⁇ -alanine, zinc N- lauroyl- ⁇ -alanine, calcium N-lauroyl-glycine etc.; amide sulfonic acid polyvalent metal salt such as calcium N-Iauroyl-taurine, calcium N-palmitoyl-taurine; N-acyl basic amino acid such as N ⁇ -lauroyl-L-lysine, N ⁇ -palmitoyl-lysine, N ⁇ -palmitoyl ornitine, N ⁇ -lauroyl arginine, hardened lanolin fatty acid acyl arginine and the like; N-acylpolypeptide such as N-lauroylglycyl glycine; ⁇ -amin
  • Preferred ultraviolet absorbing agents can comprise paraaminobenzoic acid, paraamionoethyl benzoate, paraamino amyl benzoate, paraamino octyl benzoate, ethyleneglycol salicylate, phenyl salicylate, octyl salicylate, benzyl salicylate, butylphenyl salicylate, homomentyl salicylate, benzyl cinnamic acid, paramethoxy 2-ethoxy ethyl cinnamic acid, paramethoxy octyl cinnamic acid, diparamethoxy mono-2-ethylhexane glyceryl cinnamic acid, paramethoxy isopropyl cinnamic acid, diisopropyl-diisopropyl cinnamate ester mixture, urokanic acid, ethyl urokanic acid, hydroxy methoxy benzophen
  • Preferred preservatives can comprise hinokitiol, trichloric acid, trichlorohydroxydiphenylether, chlorohexidine glucuronate, phenoxyethanol, resorcine, isopropylmethylphenol, azulene, salicylic acid, zinc pilithione, benzalconium HCl, photosensitizer 301, mononitroguaiacol Na, undecylenic acid etc.
  • Preferred antioxidants can comprise butylhydroxyanisole, propyl gallate, ellisorbate and the like.
  • Preferred pH controllers can comprise citric acid, sodium citrate, malic acid, sodium malate, fumaric acid, sodium fumaric acid, succinic acid, sodium succinic acid, sodium hydroxide, sodium hydrogen phosphate and the like.
  • Preferred alcohols can comprise cetyl alcohol etc.
  • the amount of the other ingredients ranges from 0.01 to 5%, more preferably, 0.01 to 3% in that of total composition.
  • ingredients such as water-soluble vitamin, lipid soluble vitamin, peptide polymer, polysaccharide polymer, sphingolipid, seaweed extract and other ingredients, can be obtained by conventional methods disclosed in the literature (e.g., see
  • the hardy kiwifruit preparations of the present invention can be used safely. They are non-toxic to animals and exhibit no substantial adverse effects.
  • any of the above-identified compositions can be formulated with the hardy kiwifruit preparations of the present invention (including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or fraction thereof), using any suitable dose or amount of the hardy kiwifruit preparation that is sufficient to achieve the desired biological activity for the hardy kiwifruit preparation as described above, when administered one or more times over a suitable time period.
  • suitable amounts or doses can vary depending upon the goal of the administration or the condition or the disease being treated, and also on the weight of the subject, severity, drug form, route and period of administration, and may be chosen- by those skilled in the art.
  • a suitable amount or dose of the hardy kiwifruit preparation of the present invention can include, in one embodiment, an amount of from about 0.1 g to about 1Og per kg body weight of the patient, and preferably, from about 1 to 3 g per kg by weight/day of the inventive hardy kiwifruit or extract of the present invention.
  • the dose may be administered once per day, several times per day, or in longer increments (e.g., every few days, weekly, monthly, etc.), as desired.
  • the amount of hardy kiwifruit preparation of the present invention should be present between about 0.01% to 100% by weight, and preferably between about 0.01% and about 95% by weight, and more preferably 0.5 to 80% by weight based on the total weight of the composition, including any amount in between 0.01% and 100%, in 0.01% increments.
  • a pharmaceutical composition of the present invention can contain about 0.01- 50% by weight of the hardy kiwifruit preparation of the present invention based on the total weight of the composition.
  • an extract or other preparation of the hardy kiwifruit preparation of the present invention may be provided in any composition at from about 20% to 90% highly concentrated liquid, powder, or granule, including any increment between 20% and 90%, in 1% increments.
  • a cosmetic composition comprises the hardy kiwifruit preparation of the present invention in an amount of from about with 0.01 to 30%, and more preferably, 0.01 to 5% by the weight based on the total weight of the composition, including any increment between 0.01% and 30%, in 0.01% increments.
  • a food additive or a beverage when a composition comprising the hardy kiwifruit preparation of the present invention that is added to food, a food additive or a beverage, can be provided in an amount ranging from about 0.1 to 95 w/w %, preferably 1 to 80 w/w % of total weight of food, additive or beverage, including any increment between 0.1 and 95 w/w%, in 0.1 w/w% increments, or about 1 to 30 g per 100 ml and preferably 3 to 10 g per 100 ml, including any increment between 1 g per 100ml and 30g per 100 ml, in 1 g increments, of a health beverage composition.
  • a health food of the present invention comprises the hardy kiwifruit preparation of the present invention as 0.01 to 80%, preferably 1 to 50% by weight based on the total weight of the composition, including any increment between 0.01% and 80%, in 0.1 % increments.
  • a health food beverage comprises the hardy kiwifruit preparation of the present invention in an amount of from about 0.01 to about 20% by weight of the total weight of the composition, including any increment between 0.01% and 20%, in 0.01% increments. Additional components may include: amino acids 0.001 to 5% by weight, vitamins 0.001 to 2% by weight, sugars 0.001 to 20% by weight, organic acids 0.001 to 10% by weight, sweetener and flavors of a suitable amount. Providing that the health beverage composition of present invention contains the above-described hardy kiwifruit preparation of the present invention as an essential component, there is no particular limitation on the other liquid components, wherein the other component can be various sweeteners and/or flavor enhancers, such as may be added to a conventional beverage.
  • sweeteners or flavor enhancers include, but are not limited to, conventional or reduced calorie sweeteners, including monosaccharides such as glucose, fructose etc; disaccharides such as maltose, sucrose etc; conventional sugars such as dextrin, cyclodextrin; and sugar alcohols such as xylitol, and erythritol etc.
  • Additional sweeteners include natural sweeteners such as taumatin, stevia extract, levaudioside A, glycyrrhizin and derivatives thereof, and reduced calorie sweeteners such as saccharin, sucralose, aspartame and derivatives thereof.
  • the amount of the above-described sweeteners or flavor enhancers generally ranges from about 1 to 20 g, and preferably 5 to 12 g in the ratio of 100 ml of the beverage composition, including any increment between Ig and 2Og per 100ml, in Ig increments.
  • a food additive can be added to a food by deposition, spray, or mixing.
  • the amount of the additive with respect to the total composition may generally range from about 0.01 to 20 w/w % per 100 w/w % of the present composition, including any increment between 20 w/w % and 100w/w%, in 1 w/w% increments.
  • Food additives can also be mixed with a feed, such as an animal feed, in an amount of from about 5 to 100 g per 1 kg by weight based on the total dried weight of the feed, including any increment between 5g and lOOg per lkg by weight, in Ig increments.
  • a composition including a pharmaceutical composition, a nutraceutical composition, a food additive, a health food (including a beverage or a food material), or a cosmetic composition, comprising, consisting essentially of, or consisting of, any of the hardy kiwifruit preparations described herein (including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof), and in one embodiment, a crude extract, a total water-soluble extract, or an ethyl acetate extract of the hardy kiwifruit, as an active ingredient.
  • a pharmaceutical composition including a pharmaceutical composition, a nutraceutical composition, a food additive, a health food (including a beverage or a food material), or a cosmetic composition, comprising, consisting essentially of, or consisting of, any of the hardy kiwifruit preparations described here
  • compositions of the invention are described in detail above.
  • the administration or provision of the composition results in at least one of the following biological activities: (a) reduces the number of IgE-producing B cells in a patient; (b) reduces the amount of IgE produced in a patient (e.g., in the serum or plasma); (c) decreases production and/or levels of at least one Th2 cytokine (e.g., IL-4, IL- 5, IL-10); (d) increases the level of at least one ThI cytokine (e.g., IL-12, IFN- ⁇ ); (e) decreases the level of expression of the transcription factor, GAT A-3; (f) increases the level of expression of the transcription factor T-bet; (g) increases the level of expression of the transcription factor NFATc2; (h) increases the number of IgG2a-producing B cells in a patient; (i) increases the amount of IgG2a produced in a patient; (j) enhanced production or activity of ThI T lymph
  • a preferred method of the present invention includes a method to reduce leukotriene production in a patient, thereby treating or ameliorating at least one symptom of a condition or disease associated with leukotrienes in a patient.
  • the method comprises administering to said mammal an effective amount of any of the hardy kiwifruit preparations described herein (including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof), and in one embodiment, a crude extract, a total water- soluble extract, or an ethyl acetate extract of the hardy kiwifruit, together with a pharmaceutically acceptable carrier thereof.
  • the administration of the composition of the invention results in the reduction of leukotriene production or levels in the patient.
  • Diseases and conditions associated with leukotrienes include, but are not limited to, asthma, food allergy, allergic rhinitis, chronic urticaria, and allergic dermatitis.
  • preferred routes of administration include oral, inhaled and topical administration, in addition to systemic routes of administration.
  • the method described above can be used for the prevention and/or treatment of any disease or condition in which regulation of the immune response in the manner described herein would be, or could be predicted to be, beneficial to a patient.
  • any of the hardy kiwifruit preparations described herein including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate
  • allergic diseases can include, but are not limited to, asthma, allergic bronchopulmonary aspergillosis, allergic bronchitis bronchiectasis, hypersensitivity pneumonitis, allergic sinusitis, anaphylaxis, allergic rhinitis, allergic conjunctivitis, allergic dermatitis, atopic dermatitis, contagious dermatitis, chronic urticaria, insect allergies, food allergies and drug allergies.
  • the allergic disease is atopic dermatitis.
  • the patient in addition to administration of the composition comprising, consisting essentially of or consisting of a hardy kiwifruit preparation of the invention, the patient is concomitantly treated with a conventional therapy for atopic dermatitis, including but not limited to, a topical steroid medication.
  • the hardy kiwifruit preparation is administered most preferably by oral or topical administration, although the invention is not limited to such routes of administration.
  • the allergic disease is asthma.
  • the patient in addition to administration of the composition comprising, consisting essentially of or consisting of a hardy kiwifruit preparation of the invention, the patient is concomitantly treated with a conventional therapy for asthma, including but not limited to, an inhaled steroid medication or other asthma controller.
  • a conventional therapy for asthma including but not limited to, an inhaled steroid medication or other asthma controller.
  • the hardy kiwifruit preparation is administered most preferably by oral or inhaled administration, although the invention is not limited to such routes of administration.
  • non-allergic skin inflammation diseases can include, but are not limited to, various skin troubles caused by inflammation such as pimples, acne and the like.
  • the above-described cosmetic compositions comprising any of the hardy kiwifruit preparations described herein (including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof), and in one embodiment, a crude extract, a total water-soluble extract, or an ethyl acetate extract of the hardy kiwifruit, are useful for preventing, treating, and/or improving skin inflammation in a patient.
  • non-allergic inflammatory diseases that can be prevented or treated using the compositions and methods described herein include, but are not limited to, various dermatitis conditions, systemic lupus erythematosus (SLE), retinal inflammation, gastritis, retinopathy, hepatitis, enteritis, pancreatitis, nephritis and similar conditions where reduction of a Th2 type immune response and/or enhancement of a ThI type immune response would be beneficial.
  • SLE systemic lupus erythematosus
  • any of the hardy kiwifruit preparations described herein including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract or concentrate thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or concentrate or fraction thereof
  • Preferred viruses from which to protect a mammal by preventing or treating a viral infection include, but are not limited to, Coxsackie viruses, cytomegaloviruses, Epstein-Barr viruses, flaviviruses, hepatitis viruses, herpes viruses, influenza viruses, measles viruses, mumps viruses, papilloma viruses, parainfluenza viruses, parvoviruses, rabies viruses, respiratory syncytial viruses, retroviruses and varicella viruses.
  • lymphotrophic viruses from which to protect a mammal by preventing or treating a viral infection include T-lymphotrophic viruses, such as human T-cell lymphotrophic viruses (HTLVs, such as HTLV-I and HTLV-II), bovine leukemia viruses (BLVs) and feline leukemia viruses (FLVs).
  • T-lymphotrophic viruses such as human T-cell lymphotrophic viruses (HTLVs, such as HTLV-I and HTLV-II), bovine leukemia viruses (BLVs) and feline leukemia viruses (FLVs).
  • lentiviruses include human (HIV), simian (SIV), feline (FIV) and canine (CIV) immunodeficiency viruses, with HIV-I and HIV-2 being even more preferred.
  • any of the hardy kiwifruit preparations described herein including any part of the fruit, the whole fruit, the stem, the leaf, the bark, or the root, and including any preparation or extract thereof, including dried preparations, non-extracted but processed preparations, fresh fruit, fruit juice, or any extract or fraction thereof
  • Cancers to be treated or prevented using the methods and compositions of the present invention include, but are not limited to, melanomas, squamous cell carcinoma, breast cancers, head and neck carcinomas, thyroid carcinomas, soft tissue sarcomas, bone sarcomas, testicular cancers, prostatic cancers, ovarian cancers, bladder cancers, skin cancers, brain cancers, angiosarcomas, hemangiosarcomas, mast cell tumors, primary hepatic cancers, lung cancers, pancreatic cancers, gastrointestinal cancers, renal cell carcinomas, hematopoietic neoplasias, and metastatic cancers thereof.
  • the hardy kiwifruit preparation can be administered in conjunction with another therapy or composition that is useful for treating the particular condition.
  • the hardy kiwifruit can be considered to be an adjunct to a conventional therapy, to enhance the improvement, recovery, or amelioration of symptoms in the patient.
  • Particularly preferred types of conventional agents or therapies that can be used together with a hardy kiwifruit preparation of the invention include, but are not limited to, steroids (including corticosteroids, and including oral, inhaled and injected), antihistamines (any type, including systemic, topical, inhaled), antibodies (e.g., anti-IgE, anti-IL-10), antibiotics, cyclosporins, antimycotics, respiratory function controllers, analgesics, ⁇ -agonists (long or short acting), leukotriene modifiers (inhibitors or receptor antagonists), cytokine or cytokine receptor antagonists, phosphodiesterase inhibitors, sodium cromoglycate, nedocrimil, caffeine, theophylline, carbobenzoxy beta-alanyl taurine, inhibitors of T cell function and other anti-inflammatory agents.
  • steroids including corticosteroids, and including oral, inhaled and injected
  • antihistamines any type, including systemic, topical, inha
  • compositions can be administered or provided to any member of the Vertebrate class, Mammalia, including, without limitation, primates, rodents, livestock, horses and domestic pets.
  • Preferred patients to protect are domestic pets (e.g., dogs, cats) and humans, with humans being particularly preferred.
  • AU modes of administration are contemplated.
  • the terms "patient”, “subject” and “individual” can be used interchangeably.
  • Administration routes include in vivo, in vitro and ex vivo routes. Ex vivo refers to performing part of the regulatory step outside of the patient. In vivo routes include, but are not limited to, intravenous administration, intraperitoneal administration, intramuscular administration, intranodal administration, intracoronary administration, intraarterial administration (e.g., into a carotid artery), subcutaneous administration, transdermal delivery, intratracheal administration, intraarticular administration, intraventricular administration, inhalation (e.g., aerosol), intracranial, intraspinal, intraocular, aural, intranasal, oral, pulmonary administration, impregnation of a catheter, intracutaneous, intrathecal, epidural, intracerebroventricular injection, and direct injection into a tissue.
  • intravenous administration intraperitoneal administration, intramuscular administration, intranodal administration, intracoronary administration, intraarterial administration (e.g., into a carotid artery), subcutaneous administration
  • a composition is administered by a parenteral route (e.g., subcutaneous, intradermal, intravenous, intramuscular and intraperitoneal routes).
  • Intravenous, intraperitoneal, intradermal, subcutaneous and intramuscular administrations can be performed using methods standard in the art.
  • Aural delivery can include ear drops
  • intranasal delivery can include nose drops or intranasal injection
  • intraocular delivery can include eye drops.
  • Aerosol (inhalation) delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad. ScL USA 189:11277-11281, 1992, which is incorporated herein by reference in its entirety).
  • a composition or vaccine of the invention can be formulated into a composition suitable for nebulized delivery using a suitable inhalation device or nebulizer.
  • Oral delivery can be performed by complexing a composition of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal, for example, as tablets or capsules, as well as being formulated into food and beverage products. Examples of such carriers, include plastic capsules or tablets, such as those known in the art.
  • Direct injection techniques are particularly useful for site- specific administration of a compound.
  • Oral delivery or topical delivery are particularly preferred routes of delivery or administration according to the present invention. Routes of administration that modulate mucosal immunity are useful in the treatment of viral infections and some allergic conditions. Such routes include bronchial, intradermal, intramuscular, intranasal, other inhalatory, rectal, subcutaneous, topical, transdermal, vaginal and urethral routes.
  • mice BALB/c female mice (6 wks old) were obtained from Daehan Biolink, Co. Ltd. (Korea), kept in an air-conditioned and pathogen-free room and acclimated for at least 1 wk. All experimental procedures mentioned below were performed in accordance with the institutional animal care and use guidelines of the Animal Experimental Center at Seoul National University.
  • PG102T dissolved in DW was extracted successively with chloroform, ethyl acetate and w-butanol resulting in PG102C, PG102E, and PG102B, respectively.
  • the remaining aqueous layer was called PGl 02W.
  • Each solvent-soluble fraction and the final aqueous residue were filtered, concentrated, freeze-dried, and dissolved at concentrations of 100 mg/ml. All preparations were stored at -80 0 C until such time as needed.
  • Bioassay in U266B1 cells The IgE inhibitory effects in LPS-stimulated human B lymphoblastoma cells, U266B1 (ATCC, Manassas, VA), were measured as described by Kim et al.
  • Splenocytes were incubated with OVA at 100 ⁇ g/ml in the presence of PG102T (1 mg/ml), PG102C, PG102E, PG102B, PG102W (all at 0.1 mg/ml) or media as a control for 3 days. Following incubation, the culture supernatants were collected to detect the level of cytokines (IL-4, IL-5, IL-12 and IFN- ⁇ ) using ELISA kits (Endogen, Cambridge, MA). The virtually identical procedure was used to determine the specific activity of PG102T and PGl 02E.
  • OVA-sensitized mice were orally treated with PG102T (15 mg/kg/day) or PG102E (1.5 mg/kg/day) with dexamethasone (DEX, 0.5 mg/kg/ day) or DW (100 ⁇ L/mouse/day) as a control, once a day from day 14 to day 24. Naive mice were orally treated with DW.
  • PG102T 15 mg/kg/day
  • PG102E 1.5 mg/kg/day
  • DEX dexamethasone
  • DW 100 ⁇ L/mouse/day
  • mice On day 21, blood was obtained from individual mice by eye-bleeding and isolated plasma samples were kept at - 80°C until time of use.
  • the level of total IgE was measured by way of a mouse IgE detection kit (Shibayagi, Gunma, Japan).
  • the levels of total IgG subtypes and OVA-specific Ig isotypes were determined by the sandwich ELISA method (Hirano et al., J Immunol Methods 1989; 119:145-50).
  • splenocytes were prepared from animals on day 24, resuspended in culture medium (RPMI- 1640 containing 10% FBS), seeded onto a 24 well plate (5x 10 6 cells/ml/well) and incubated with OVA only at 100 ⁇ g/ml for 3 days.
  • Splenocytes isolated from naive mice were cultured in the absence of OVA.
  • the levels of IL-4, IL-5, IL-10, IL-12, IL-13 and IFN- ⁇ in supernatants were detected by ELISA (Endogen and R&D Systems, Minneapolis, MN).
  • Splenocytes were exposed to GolgiStop (PharMingen, San Diego, CA) as an intracellular protein transport inhibitor for 4 h and prepared for the detection of IL-4 or IFN- ⁇ -producing cells.
  • Cells were fixed, permeabilized, and incubated with PE or FITC-conjugated antibodies specific to mice CD4, IL-4 or IFN- ⁇ as described by Kyoko et al. (Kyoko et al., J Derm Science 2002;29: 19-25).
  • IgE products cells were incubated with PE-conjugated anti-mouse CD 19, followed by FITC- conjugated anti-mouse IgE (all from PharMingen).
  • splenocytes were cultured with OVA on cover slips for 2 days, fixed, permeabilized, and stained using FITC- conjugated anti-mouse IgE and PE-conjugated anti-mouse CD 19, and finally observed by the MRC- 1024 Laser Scanning Confocal Image System (Bio-Rad Laboratories Inc., Hercules, CA) as described by Semper et al. (Semper et al., J Allergy CHn Immunol 2003;l 12:141-9). Western blotting. Splenocytes from the individual group of mice were cultured with
  • OVA OVA for 2 days, collected (10 7 cells/group), and then lysed for the preparation of protein samples. Immunoblotting was performed using antibodies specific to mouse GAT A-3, T- bet, NFATc2 (Santa Cruz Biotechnology, Santa Cruz, CA) or ⁇ -actin (Sigma) as a loading control.
  • RNA preparation and quantitative real-time PCR Total RNA was isolated from splenocytes cultured with OVA for 2 days using TRIzol Reagent (GIBCOBRL, Carlsbad, CA). Isolated total RNA was used in reverse transcription (RT)-PCR by the AMV RT System (Roche, Mannheim, Germany), followed by quantitative real-time PCR using the ABI PRISM 7700 Sequence Detection System (Applied Biosystems, Foster City, CA).
  • NFATc2 forward 5'-GCACATAAGGCCATCAGCTCA-S' (SEQ ID NO:5) reverse 5'-TCGCCAGAGAGACTGGCAA-S ' (SEQ ID NO:6)
  • mice were immunized with OVA on day 0 and boosted on day 14. Ten days later, mice were sacrificed and spleens were taken to isolate splenocytes. Splenocytes derived from OVA-sensitized mice were stimulated with OVA and cultured for 3 days in the presence of five preparations of PG102. As shown in Table I, when cells were grown in the presence of OVA, the levels of IFN- ⁇ , IL-4 and IL-5 were increased to a hundred picograms or nanograms.
  • IL-12 In the case of IL-12, the background level was high, but OVA stimulation decreased the level by approximately 3-fold to 415 ⁇ 48 pg/ml. Treatment with PG102T resulted in an almost 150% increase in the level of IL-12. IFN- ⁇ behaved differently. Splenocytes from na ⁇ ve animals produced undetectable levels of IFN- ⁇ , but OVA- stimulated splenocytes secreted almost 2.5 ng/ml. Treatment with PGl 02T decreased the level of IFN- ⁇ by 40%. When OVA-stimulated splenocytes were grown in the presence of PGl 02T, the levels of IL-4 and IL-5 were decreased by 62% and 39%, respectively.
  • PGl 02E which showed the highest inhibitory effect on IgE production in the above experiment, also reduced the levels of IL-4 and IL-5 by 78% and 59%, while inducing the level of IL- 12 by 220%. In contrast, PGl 02C reduced the level of all cytokines measured in this study due to its cytotoxic effect on splenocytes.
  • Both PGl 02B and PGl 02W increased the level of IL- 12 and decreased that of IFN- ⁇ , IL-4 and IL-5, but not in a statistically significant manner.
  • PG102T and PG102E might contain the compound(s) that inhibits the production of IgE and controls the expression of selective ThI and Th2 cytokines.
  • the inventors chose to use two preparations, PGl 02T and PGl 02E, for further in vivo studies. Determination of specific activity of PGl 02T and PG102E. Based on their pleiotropic activities on the immune system, PG102T and PGl 02E as derived from a plant source were thought to contain more than one active compound.
  • the inventors developed a reliable bioassay based on the inhibitory activity of PGl 02 on the production of IL-4 in OVA-stimulated splenocytes as described above.
  • the level of IL-4 was induced from an undetectable level to a few hundred picograms.
  • the production of IL-4 was inhibited in a dose-dependent manner (Fig. 2).
  • neither PGl 02 T nor PG102E demonstrated any cytotoxic effects.
  • the concentration of PG102T with 50% inhibitory activity was 806.9 ⁇ g/ml and that of PG102E was 91.8 ⁇ g/ml.
  • the IC 50 value of each preparation was defined as one activity unit, and total units were obtained.
  • PGl 02T and PGl 02E contained specific activity of 1.2 units/mg and 10.9 units/mg, respectively. This method was used for the quality control of experimental samples from A arguta. Effects of PG102T and PGl 02E on production of ThI and Th2 cytokines in the
  • OVA-s ensitized murine model OVA-s ensitized murine model.
  • effects of PG102T and PG102E were tested on the production of various cytokines involved in the modulation of ThI and Th2 pathways using the OVA-sensitized murine model.
  • mice untreated with OVA were orally fed with DW.
  • concentrations of PG102T and PGl 02E used in these experiments are the minimal dosage having maximal activity as per the preliminary dose-response experiment.
  • mice were sacrificed, and spleens were isolated to prepare splenocytes. Splenocytes from each group of mice were incubated in the presence of OVA for 3 days, and cultured supernatants were collected to measure the level of cytokines (Table II). Compared with na ⁇ ve splenocytes, the level of IL- 12 was decreased in DW-treated mice.
  • oral administrations of PGl 02T and PG102E increased its level by 1.7- and 2.6-fold, respectively.
  • the level of IFN- ⁇ was enhanced by OVA-stimulation, and PGl 02T or PGl 02E further increased the level of IFN- ⁇ . Contrary to PG102T and PG102E, DEX suppressed the level of both IL-12 and IFN- ⁇ .
  • Th2 cytokines tested in this study was highly increased in OVA- stimulated splenocytes.
  • treatment with PGl 02T suppressed OVA-mediated overproduction of IL-4, IL-5, and IL-10 by 44%, 32% and 44%, respectively.
  • PGl 02E also inhibited the level of these three cytokines by 64%, 69% and 51%, respectively.
  • DEX also lowered the concentration of all three cytokines.
  • IL-13 was decreased by either PG102 or DEX, but not in a statistically significant manner.
  • PGl 02T and PGl 02E appear to have distinctive biological activities that can differentially modulate the production of ThI and Th2 cytokines.
  • PGl 02T or PG102E The capability of PGl 02T or PG102E to down-regulate the level of total IgE in plasma was comparable to that of DEX.
  • administration of both PG102T and PG102E decreased the level of Th2-mediated IgGl, whereas the level of Thl-mediated IgG2a was highly elevated in a statistically significant manner.
  • the level of IgG2b was not significantly affected in all situations (Table IIIA).
  • IgE ng/ml
  • IgGl ⁇ g/ml
  • IgG2a ⁇ g/ml
  • IgG2b ⁇ g/ml
  • OVA-specific Ig (% control of OD)
  • CD4+IFN- ⁇ + cells were slightly increased from 7.2% to 9.2 or 9.6% when animals were treated with PG102T or PGl 02E, though not in a statistically significant manner. There was little change in the number of both cell types in DEX-administered animals. These results indicated that PGl 02T and PGl 02E might regulate the proportion of IL-4- and IFN- ⁇ -producing T cells.
  • the inventors analyzed the effects of PG102T and PGl 02E on the IgE- producing B cells. Compared with the DW treatment, oral administration of PGl 02T or PG102E resulted in a decrease of about 40% in the number of CD19+IgE+ B cells.
  • PG102T and PG102E were determined using a quantitative real-time PCR technique (Fig. 4B).
  • PGl 02T treatment decreased the mRNA level of GATA-3 by almost 3-fold, whereas it increased that of T-bet and NFATc2 by approximately 14-fold and 2-fold, respectively.
  • PG102E which showed higher biological activity in terms of IgE inhibiting effect, changed the levels of GAT A3, T-bet and NFATc2 to a lesser extent than PGl 02T.
  • DEX lowered the levels of both GAT A3 and NFATc2, and slightly elevated that of T-bet.
  • PG102T and PG102E The effects of PG102T and PG102E on cellular transcription factors might explain how two preparations work at the molecular level. Both PG102T and PG102E decreased the level of GATA-3, while increasing that of T-bet and NFATc2. GATA-3 is known to strongly trans-activate the IL-5 promoter and the IL-4 enhancer element (Lee et al., J Exp Med 2000;192:105-15; Ting et al., Nature 1996;384:474-8).
  • T-bet is involved in the commitment of ThI cells by inducing the synthesis of IFN- ⁇ in ThI cells (Lighvani et al., Proc Natl Acad Sd USA 2001;98:15137-42; Szabo et al., Cell 2000;100:655-69). In recent studies, it was also reported that T-bet regulates the isotype-switching to IgG2a and the production of IFN- ⁇ in B cells (Gerth et al., hit Immunol 2003;15:937-44).
  • NFATc2 is a non-selectively expressed antigen-inducible transcription factor. Although this protein binds to the enhancer of IL-4 and the IFN- ⁇ promoter in stimulated Th cells, it has been assumed that NFATc2 plays a more important role in driving na ⁇ ve T cells to effector ThI cells.
  • T cells from NFATc2-/- mice secreted much higher levels of IL-4 than wild type T cells (Kiani et al., Blood 2001;98: 1480-8; Erb et al., Infect Immun. 2003;71(l l):6641-7).
  • the present study showed that PGl 02T or PGl 02E increased the level of NFATc2 expression.
  • PG102T or PG102E favor ThI responses and suppress IgE biosynthesis
  • PGl 02T or PG102E acts on antigen presenting cells, which play a key role in Th cell differentiation through the secretion of soluble factors including IL- 12 and the expression of co- stimulatory molecules such as B7-1 (ThI) and B7-2 (Th2) (Kuchroo et al., Cell 1995;80:707-18).
  • B7-1 B7-1
  • Th2 B7-2
  • Another possibility is that it works directly on the differentiation process of Th progenitor cells or on the regulation of IgE-producing B cells.
  • the regulatory effects of PGl 02T or PGl 02E on ThI and Th2 systems may be due to multiple biologically active compounds.
  • the present inventors devised the bioassay system based on their effect on the production of IL-4, a major inducer of Ig isotype switching to IgE, in OVA-stimulated splenocytes.
  • This assay was sensitive and reproducible enough to calculate the specific activity of PG 102 and use it to study various factors influencing biological activities of PG 102, such as harvest time and different geographic locations for A. arguta.
  • This IL-4- based assay is now being used for the purification of active compound(s) as well as the quality control of PG102 reagents used in a variety of experiments.
  • the inventors' toxicity data indicate that PG102T is safe, hi repeated dose toxicity trials, oral administration of PG102T (500, 1000, 2000 mg/kg/day) for 4-12 weeks produced no adverse effects in rats. These results are in stark contrast with the data obtained from mice administered DEX (0.5mg/kg/day), which showed severe reductions in body weights and mass of spleens (data not shown). These data, describing the various biological activities of PG102T and PG102E, indicate that extracts of A. arguta are safe, natural preparations, with a minimum risk of side effects, for potential use in the treatment of various allergic disorders.
  • Example 2 The following example demonstrates that at least two specific extracts prepared from A. arguta, denoted PG102T and PG102E, have a therapeutic effect on atopic dermatitis.
  • NC/Nga mice were established as an inbred strain in 1957 based on Japanese fancy mice (Nishiki-Nezumi). When kept under specific pathogen-free (SPF) conditions, mice remain normal and healthy. However, when placed in conventional surroundings, clinical signs begin with scratching behavior initiating from the age of 8 weeks followed by the onset of the eczematous condition. The promptly developed eczema is typically localized on the face, ears, neck and back region. The affected mice display the various clinical signs including hemorrhage, superficial erosion, deep excoriation, scaling, dryness of the skin, and growth retardation (Hiroshi et al., Int Immunol 1997;9(3):461-466).
  • the infiltration of numerous CD4+ T cells and eosinophils and the increased number of mast cells with degranulation are observed.
  • the plasma level of IgE is markedly elevated from the age of 8 weeks, coinciding with the appearance of the skin lesions.
  • the infiltrated cells in the skin lesions express IL-4, IL-5 and TARC, but little or no IFN- ⁇ , resulting in manifestation of Th2-dominant immune reactions (Masayuki et al., Int Ach Allergy Immunol 2003;132:355-63; Christian et al., MoI Med Today 2000;5:209-10).
  • NC/Nga mouse can be an excellent animal model for human AD and also that the modulation of Thl/Th2 and the suppression of IgE biosynthesis may be a therapeutic strategy that can fundamentally improve the clinical symptoms of AD in both humans and mice.
  • PG102T and PG102E used in this study were prepared from A. arguta as described previously (Park et al., J. Allergy Clin. Immunol., 116:1151-1157, 2005 and Example 1).
  • SPF NC mice as a negative control, received DW.
  • a total clinical severity score for AD-like lesions was defined as the sum of the individual scores graded as 0 (none), 1 (mild), 2 (moderate) and 3 (severe) for each of five signs and symptoms (itch, erythema/hemorrhage, edema, excoriation/erosion and scaling/dryness). Measurement of plasma level of immunoglobulins, cytokines, and chemokine.
  • Spontaneously induced allergic responses were monitored by measuring the plasma levels of immunoglobulins including IgE and IgG2a, cytokines, and chemokines.
  • Blood was collected from the retro-orbital plexus with glass capillary tubes at the ages of 7, 10, 12, and 14 weeks, and separated plasma samples were stored at -8O 0 C until use.
  • the level of IgE was determined by a mouse IgE detection kit (Shibayagi, Gunma, Japan) and that of IgG2a was measured by the sandwich ELISA method as described by Hirano et al. (Hirano et al., J Immunol Methods 1989; 119: 145-50).
  • mice IL-4, IL- 12, eotaxin and TARC were also measured by ELISA kits (Endogen, Cambridge, MA and R&D Systems, Minneapolis, MN) according to the manufacturer's instructions.
  • ThI /Th2 cytokine production by splenocytes derived from NC/Nga mice At the age of 14 weeks, conventional NC mice orally treated with PG102T, PG102E, DEX or DW and SPF NC mice were sacrificed by decapitation.
  • spleens of an individual group were obtained, and splenocytes isolated from spleens were resuspended in culture medium (RPMI- 1640 containing with 10% heat- inactivated FBS). Splenocyte suspension was seeded to a 24 well culture plate, and the final concentration was adjusted to 5x10 6 cells/ml /well.
  • the thickness of both the epidermis and dermis were measured as the distance from the stratum corneum of the epidermis to the basement membrane of the dermis. The distance was expressed as the mean of three random fields for which 5 measurements were averaged.
  • cytokine and chemokine expression in the skin biopsies The levels of IL-4, IL-5, eotaxin and TARC in the skin biopsies from the face were measured by ELISA. Briefly, the tissue from facial skin lesions was excised, homogenized in lysis buffer, and then the freezing/thawing procedure was repeated three times. After centrifugation, the supernatants containing total cellular protein were quantified and used to detect the level of cytokines and chemokines. Results were normalized to the total amount of protein prepared from tissue lysates.
  • NC mice were orally administered PG102T [50 mg (60 units)/kg/day], PGl 02E [5 mg (54.5 units) /kg/day], DEX (2.5 mg/kg/day) or DW (100 ⁇ L/mouse/day ) on a daily basis for 7 weeks and the progression of atopic dermatitis was observed.
  • the dosage of PG102T and PG102E was based on the concentration that gave therapeutic effects in the OVA-sensitized murine model used in the previous experiments (see Example 1 and Park et al., J. Allergy Clin. Immunol., 116:1151-1157, 2005).
  • PG102T and PG102E decrease the production of IgE and IgGl, while they increase that ofIgG2a in plasma.
  • conventional NC mice also show the elevated level of IgE in plasma after the onset of dermatitis. Therefore, it was tested whether oral administration of PG102T or PG102E could control the plasma level of Th2-mediated IgE and IgGl and Thl-mediated IgG2a. From 7 weeks of age, animals were orally fed with PGl 02T, PGl 02E, DEX or DW on a daily basis, and blood samples were obtained at the age of 7, 10, 12 and 14 weeks.
  • NC mice Under SPF conditions, NC mice normally produced approximately 150 ng/ml of total IgE, but when animals were placed under conventional conditions, IgE levels gradually increased with aging, to almost 17 ⁇ g/ml at 14 weeks of age.
  • Administration of PG102T or PGl 02E lessened the plasma level of IgE from 10 weeks of age in a statistically significant manner, resulting in a 5 -fold lower level of IgE at the end of the experiment.
  • the IgE- lowering effect of PG102T and PGl 02E was comparable to that of DEX used as a positive control (Fig. 6A).
  • the level of IgGl another Th2-mediated Ig class, was measured.
  • DW-treated mice produced a level of IgGl greater than 5 mg/ml.
  • administration of PG102T and PGl 02E decreased its level by 75% and 90%, respectively.
  • the level of IgG2a which belongs to the Thl-mediated Ig class, was increased by approximately 180% in conventional NC mice treated with PGl 02T.
  • PG102E also induced the level of IgG2a in plasma (Fig. 6B).
  • PG102T or PGl 02E may regulate the balance of ThI /Th2 cytokine production in plasma and splenocytes.
  • the levels of IL-4 and IL- 12, representing the Th2 and ThI pathways, respectively, were measured in plasma at 12 weeks of age. Compared with conventional NC mice treated with DW, the level of IL-4 was lowered in mice treated with PGl 02T or PGl 02E by 60% and 76%, respectively (Table IVA). Oral administration of PGl 02T or PGl 02E elevated the level of IL- 12 in a statistically significant manner.
  • the inventors also analyzed the effects of PG102T and PG102E on the production of ThI and Th2 cytokines in splenocytes isolated from NC mice. Mice were sacrificed at 14 weeks of age and spleens were obtained to isolate the splenocytes. Splenocytes from each group of mice were stimulated with a T cell-specific mitogen, ConA, for 3 days, and the levels of various cytokines were detected. In the presence of ConA, the level of all Th2 cytokines was highly elevated, but treatment with PG102T or PGl 02E reduced the levels of IL-4, IL-5 and IL-10 by 24% to 78% (Table IVB). DEX also inhibited the levels of all three Th2 cytokines, although the decrease of IL-5 by DEX was not statistically significant. The level of IL- 13 was not influenced by PGl 02 or DEX.
  • Plasma samples were isolated from each group of mice at the age of 12 weeks. All splenocytes from NC mice were stimulated with ConA during the culture. Values are expressed as means ⁇ SEM for five animals. *, P ⁇ 0.05 and **, P ⁇ 0.01, vs DW-treated mice (Student's T-test). ND, not detectable.
  • PGl 02T and PGl 02E increased the level of ThI cytokines, while decreasing that of selective Th2 cytokines, unlike DEX, which indiscriminately inhibited the expression of almost all cytokines measured in this study.
  • PGl 02 not only prevents eosinophilia, but also decreases the level of eotaxin and TARC.
  • Dermal infiltration of inflammatory cells including eosinophils is an important feature of atopic dermatitis in NC mice. Because the presence of inflammatory cells in the skin lesions may have resulted from their mobilization from bone marrow into blood, the inventors first analyzed the number of total leukocytes and eosinophils in the peripheral blood at 12 weeks of age. As shown in Fig. 7 A, the number of total leukocytes of conventional NC mice was increased upon the onset of the dermatitis.
  • the number of eosinophils was greatly increased in DW-treated mice under conventional conditions, resulting in eosinophilia.
  • PGl 02T or PGl 02E administration lowered the number of both total leukocytes and eosinophils, presumably contributing to the prevention of eosinophilia.
  • the changes in the number of circulating eosinophils may be manifested by the production of chemokines, leading to chemoattraction in response to inflammation (3-6). Therefore, the plasma levels of eotaxin and TARC, which are representative chemoattractants for eosinophils and Th2 cells, were determined.
  • mice In the conventional environment, DW-treated mice produced an increased level of eotaxin and TARC, but these levels were decreased in mice treated with PG102T or PG102E by approximately 25% to 50% (Fig. 7B). There was little change in the animals administered DEX. These results showed that PGl 02T and PG102E inhibited the production of eotaxin and TARC, resulting in the prevention of Th2-mediated eosinophilia, which generally coincides with the onset of dermatitis in NC mice.
  • PGl 02T or PGl 02E administration inhibits the infiltration of inflammatory cells in dermis and the thickening of the epidermis and dermis. Improvement of clinical skin condition and inhibition of Th2 response by PG102T and PG102E was also confirmed by the analysis of H&E stained sections at 14 weeks of age. Mice fed with DW exhibited a marked thickening of the epidermis and dermis, prominent hyperkeratosis, infiltration of inflammatory cells, and hemorrhage. Morphologic study indicated that these infiltrating cells in the dermis were eosinophils, mast cells and lymphocytes.
  • PGl 02 or PG102E reduces the expression of Th2-mediated cytokines and chemokines through the down-regulation of GATAS.
  • the levels of IL-4, IL-5, eotaxin and TARC were measured by ELISA at 14 weeks of age. In the skin from SPF NC mice, all four proteins were weakly expressed, but their levels were notably increased in conventional NC mice.
  • Administration of PGl 02T or PGl 02E lowered the levels of IL-4, eotaxin and TARC by more than 30%. The effect on IL-5 expression was more prominent, with its level being increased by almost 90%.
  • DEX inhibited the levels of IL-5 and TARC, but not that of IL-4 and eotaxin (Fig. 9A).
  • STAT6 and GATA3 were well known to play critical roles in the differentiation of Th2 cells and the production of Th2-specific cytokines and chemokines (Arakawa et al., Clin Exp Immunol 2004;135(3):505-10; Gunther et al., J Allergy CHn Immunol 2004; 113:987-94; Konishi et al., Proc Natl Acad Sd 2002;99(17): 11340-5). As shown in Fig. 9B, the level of GATA-3 protein was lowered by both PG102T and PG102E.
  • phosphorylated STAT6 (pSTAT ⁇ ) was also decreased in the conventional NC mice treated with PG102T, while it might not be the case with PGl 02E.
  • DEX suppressed the level of both GATA3 and pSTAT6.
  • PGl 02T and PGl 02E might down-regulate the level of Th2-specific cytokines and chemokines by inhibiting the expression of GATA3.
  • AD is a major allergic disease that often begins during infancy. A significant fraction of affected individuals develop asthma and/or allergic rhinitis later in life (Leung, Clin Exp Immunol 1997;107(suppl. l):25-30).
  • AD results from dermal inflammation caused by an abnormal immune response, in particular, overactivation of the Th2 pathway.
  • the inventors have discovered that PG102T and PG102E, water-soluble fractions derived from A. arguta, control the production of selective ThI- and Th2-mediated cytokines and also that of IgE in the OVA-sensitized mouse model (see Example 1). Based on these data, it was reasoned that these plant extracts might be useful for the treatment of various allergic diseases (Mayaumi et al., J Allergy CHn Immunol 2000;106:159-66; Hisae et al., Phytother Res 2001; 15:506-10).
  • PGl 02 could produce any actual therapeutic effects(s) on atopic dermatitis using NC/Nga mice as a model system.
  • the biological consequences of such biochemical changes in the NC/Nga mouse model include the highly decreased number of eosinophils in the peripheral blood as well as in the skin lesions, the suppression of the thickening of the epidermis and dermis, and the inhibition of the infiltration of various inflammatory cells.
  • Of particular interest is a decrease in the regional expression levels of eotaxin, TARC, IL-4 and IL-5.
  • the levels of these proteins are abnormally high in the skin lesions of NC/Nga mice grown under the conventional environment. When animals were orally administered PGl 02, however, these chemokines and cytokines were found to be at the virtually normal level.
  • Eotaxin together with IL-5, is known to be a potent chemoattractant for eosinophils, while TARC produced by keratinocytes (and also Th2 cells) is thought to attract Th2 cells, and induces the pathological responses typically associated with atopic dermatitis.
  • the receptors for eotaxin and TARC are CCR3 and CCR4, respectively, which are both highly expressed in Th2 cells (Christian et al., J Clin Invest 1999;104: 1097-105; Tomomi et al., J Allergy Clin Immunol 2110;107:353-8; Weilie et al., J Clin Invest 2002;109:621-8; Masayuki et al., IntAch Allergy Immunol 2003;132:355-63).
  • PGl 02 acts first on cellular transcription factors, for example GAT A-3, and subsequently on the expression of key cytokine players involved in the ThI and Th2 systems such as IL-4 and IFN- ⁇ , inducing the cascade reaction, leading to a decrease in the level of IgE and respective chemokines (Zhu et al., Nat Immunol. 2004;l 1 : 1157-65).
  • PGl 02 may initially work at the local level; for example, decreasing the level of eotaxin and TARC, inhibiting their chemoattractive functions, lowering the number of eosinophils at the systemic and local levels, and suppressing the histopathological development seen in conventionally grown NC/Nga mice.
  • the activity of PGl 02 may be due to multiple compounds acting at various levels of allergy-related biochemical pathways. Overall, PGl 02 appears to operate on key biological factors related to atopic dermatitis in this animal model, treating the condition at its source rather than simply providing relief of symptoms .
  • PG102T and PG102E were derived from an edible fruit. No toxicity was found in repeated dose toxicity experiments, in which 2000 mg/kg/day, 40-fold higher than the concentration used in this study, was administered on a daily basis for 12 weeks. Together with data from the previous experiments involving OVA-sensitized mice, the results from NC/Nga mice demonstrate that PG 102 is a safe and effective reagent for the treatment of various allergic diseases including atopic dermatitis. Given that the prevalence of atopic diseases is increasing in all major developed countries and virtually no reagent is available for the fundamental treatment of atopic dermatitis, preparations such as those described herein represent advances in the field.
  • Example 3 Example 3
  • Powdered stems (126.6 g), powdered roots (79.0 g), and finely divided bark (126.2 g) were each extracted with distilled water (1 L) at 94°C for 4h. The mixtures were then filtered, and the filtrate concentrated to dryness by rotary evaporation to provide a stem extract (9.9 g), a root extract (8.6 g), and a bark extract (2.4 g). Twenty frozen A. arguta berries (154.4 g) were thawed at room temperature, crushed, and extracted with distilled water (1 L) 91°C for 5h. The mixture was filtered, and the filtrate concentrated to produce a 'boiled' fresh fruit extract (12.8 g).
  • the juicer removed the skins from the fruit resulting in a mixture of seeds, pulp, and juice. This mixture was centrifuged (30 min., -3500 rpm) to provide 150 mL of juice. This juice was concentrated to dryness by rotary evaporation resulting in a fruit juice concentrate (24.2 g).
  • a process scale extraction of the kiwifruit was performed (Sungil Bioex Co., Ltd., Bibong, Korea). Frozen kiwifruit (1242 kg) were sliced (1/4" to 3/8" thickness) and dried in a convection dryer (65-80°C) to a moisture content of 5-20%. Batch extraction (Fig.
  • the filtrate was then concentrated under vacuum (-600 mmHg) at 55-65 0 C in an agitated stainless steel reactor equipped with an external condenser and a distillate receiver. Once the material was concentrated, it was held at 8O 0 C for an additional 30 min. to sterilize the extract.
  • the resulting material (101 kg), equivalent to PGl 02T, was designated FDOOl. Of this material, 3 kg were set aside for further testing. Good Manufacturing Practices were used throughout the process.
  • the FDOOl concentrate produced above (98 kg) was pumped to a horizontal paddle blender and mixed with an equal weight, based on the calculated solids content, of microcrystalline cellulose (MCC). Following this, the solid blend was transferred to stainless steel trays that were placed into a forced hot air dryer (70- 80 0 C) for 24 h. The dry, lumpy solids were then ground in a Fitzmill type hammer mill to produce a 40 mesh powder (118 kg).
  • This material was encapsulated (GMP Laboratories of America, Inc., Anaheim, CA) into 300 mg- or 600 mg-sized capsules, each containing a 1:1 mixture of FDOOl and MCC for use in canine and human clinical trials.
  • the following example describes in vitro testing for immunomodulating activity in A. arguta preparations.
  • the purpose of this study was to compare the relative ability of various extracts and preparations produced from A. arguta to modulate cytokine production (IL-4, IL-5, IL-IO, IL- 13, and IFN ⁇ ) in splenocyte cultures derived from ovalbumin (OVA, grade V, Sigma)- sensitized mice using ELISA (Quantikine kits, R&D systems) analysis.
  • OVA ovalbumin
  • ELISA Quantantikine kits, R&D systems
  • mice Female, Balb/c mice (Harlan, Indianapolis, IN) were sensitized by IP injection of 20 ⁇ g OVA on days 0 and 14. On day 24, following euthanasia by cervical dislocation, spleens were aseptically removed from individual mice and immediately processed for splenocyte culture development using sterile technique. The spleens were dissociated in the presence of 10 niM HEPES-buffered RPMI- 1640, by gently forcing the tissues through the grid of a 70 micron nylon mesh using the plunger from a 3 cc syringe. Large cell aggregates were removed from the resulting suspension using a FCS-gradient.
  • the splenocytes were then centrifuged (1500 rpm, 5 min.) and the resulting cell pellets were treated with RBC lysis buffer (10 min., RT) to remove the contaminating erythrocytes. The majority of the RBC lysis buffer was then removed by centrifugation (1500 rpm, 5 min.) and the pelleted splenocytes were then washed 3X with 10 mM HEPES-buffered RPMI- 1640. Following the final wash, the pelleted splenocytes were resuspended in a volume of in RPMI- 1640 containing 10% FCS and Penn/Strep (complete medium) designed to deliver a final cell density of 5 x 10 6 cells/mL. For each analysis, 5 x 10 6 splenocytes were plated into the individual wells of a 24- well plate. On day 3, the supernatants from these wells were collected and frozen in preparation for the determination of experimental results.
  • Control splenocyte cultures were also established from na ⁇ ve (non-sensitized) mice in the manner described above and plated out into the individual wells of a 24-well plate to achieve a final cell density of 5 x 10 6 cells/mL. These splenocytes were established in RPMI- 1640 containing 10% Fetal Calf Serum, Penn/Strep, and they received no additional treatment. On day 3, the supernatants from these wells were collected and frozen to serve as negative experimental controls. Stimulation of splenocyte cultures with A, arguta preparations
  • mice Ten OVA-sensitized mice were used for each preparation tested. Splenocytes from each mouse were plated (5 x 10 6 cells/mL) into 8 individual wells of a 24-well plate in complete RPMI-1640 medium containing 100 ⁇ g/mL OVA, 0.5% DMSO and either no or chosen concentrations of each of the specific test preparations under examination. 6 of the 8 wells were partitioned into 2 sets of 3 wells. Each of the wells in the sets of 3 were treated with A. arguta preparations at concentrations of either 0.25, 1.0 or 10 mg/mL. To serve as positive controls, the 7 th wells were treated with 2 ⁇ M dexamethasone (DEX), a potent glucocorticoid anti-inflammatory.
  • DEX dexamethasone
  • the 8 th wells received no additional treatment and served as an OVA-only experimental control. After 3 days of culture, the supernatants from each of the unique 8 wells per OVA-sensitized mouse were collected and frozen. These supernatants were used to determine the levels of the cytokines IL-4, IL-5, IL-IO, IL-13, and IFN- ⁇ present in the culture medium. Determination of cytokine levels in culture supernatants
  • the cytokine levels in the culture supernatants derived from the A. arguta preparation-treated splenocytes, DEX-treated splenocytes, OVA-only treated splenocytes, and untreated splenocytes from non-sensitized control mice were determined by ELISA assay. Two replicate ELISA plate wells were utilized for each cytokine level determination. Results
  • Example 5 The following example describes a comparison of in vitro activity of extracts of non- fruit parts of A. arguta, as well as alternative fruit preparations of A. arguta.
  • arguta prepared as described in Example 3; "boiled” fresh fruit preparations; the fruit juice concentrate prepared as described in Example 3; FDOOl (large scale equivalent of PG102T) prepared as described in Example 3; FDOOl powder prepared as described in Example 3 (used for clinical trials described below); a room temperature water extract of the dried A. arguta fruit; the EtOAc extract prepared as described in Example 3; and aqueous remainder, also described in Example 3.
  • the activity of three known immunosuppressive compounds, cyclosporin, dexamethasone, and quercetin were evaluated as controls. Splenocyte isolation and culturing
  • Splenocyte cells from 8 OVA-sensitized mice (8 replicates) were utilized for the analysis of each extract or preparation tested.
  • 5 x 10 6 cells splenocyte cells from each mouse were plated out into the individual wells of a 24-well plate in complete RPMI- 1640 medium containing 100 ⁇ g/mL OVA and 25 mM HEPES (pH 7.3), 1 ml per well.
  • Kiwifruit preparations were examined at concentrations of 1.0, 3.0, and 10 mg/mL. Cyclosporin, quercetin, and dexamethasone analysis
  • Splenocytes from 8 OVA-sensitized mice (8 replicates) were utilized for the analysis of each compound tested, with the exception of Quercetin where the splenocytes derived from only 2 OVA-sensitized mice were examined.
  • 5 x 10 6 CeIIs splenocytes from each mouse were plated out into the individual wells of a 24-well plate in complete RPMI- 1640 medium containing 100 ⁇ g/mL OVA and 25 mM HEPES (pH 7.3), 1 ml per well. Cyclosporin was examined at concentrations of 0.0083, 0.083, and 4.15 ⁇ M.
  • Dexamethasone was examined at concentrations of 0.01, 0.1 and 1 ⁇ M.
  • Quercetin was examined at concentrations of 1, 10, and 25 ⁇ M.
  • cytokine levels in the culture supernatants from all treatment and control wells were determined by ELISA assay. Two replicate ELISA plate wells were utilized for each cytokine level determination.
  • a greater suppression of the cytokines examined was observed as the concentrations of the A. arguta test materials were increased. In general, suppression was more pronounced against IFN ⁇ .
  • the activity of prescribed immunosuppressant compounds was similar to the A. arguta preparations in this assay.
  • the peptide cyclosporin and the glucocorticoid steroid dexamethasone exhibited potent activity ( ⁇ 1 ⁇ M) as shown in Fig. 14 A.
  • the flavonoid quercetin showed potent activity over a slightly higher concentration range (1-25 ⁇ M, Fig. 14B). Further confirmation of the ability of EtOAc to extract activity from FDOOl is demonstrated in Figs. 15A and 15B.
  • FDOOl powder which was the material used in both canine and human clinical trials (described below), was confirmed to be active in this assay as shown in Figs. 16A and 16B.
  • the objective of this study was to obtain preliminary evidence of effectiveness of FDOOl (PGl 02T), administered orally over a period of 42 days, in a small number of adult volunteers with atopic dermatitis (AD) of moderate severity. Secondary objectives of the study were to assess the tolerability and variability of response to FDOOl . Study design
  • Subjects had active, atopic dermatitis of moderate severity defined by a Physician's Global Assessment (Feldman and Krueger, Annals of the Rheumatic Diseases, 64:ii65-ii68, 2005) score of three on the severity scale of 0 to 4.
  • Subjects had AD involving a minimum of 10% of body surface area (BSA).
  • BSA body surface area
  • Subjects were currently using a topical steroid for the treatment of AD and could not be nursing or pregnant.
  • Safety and tolerability were assessed using adverse reaction reporting and standard blood chemistry, hematology and urinalysis.
  • the primary efficacy variable was the change from baseline in the Physician's Global Assessment at day 42, with analysis using the Cochran Mantel-Haenszel test (Armitage et al, Statistical Methods in Medical Research, 4th Ed., Blackwell, Oxford, 2002). Secondary variables were the day 42 changes from baseline in the signs of AD (erythema, induration, oozing/crusting and pruritus severity scores), and in total BSA as analyzed using a two-sample t-test. Descriptive statistics were presented for all baseline and post-baseline study data by treatment group on days 1, 14, 28 and 42. These statistics included sample size, means, standard deviations, frequencies, percentages, and confidence intervals, as appropriate.
  • results from subject self-assessment questionnaires were tallied and presented by treatment group. Any adverse events occurring during the study were recorded. Descriptions of adverse events included the date of onset, the date the event ended or continued, the severity of the event, the attribution, action taken, therapy taken, and the outcome. These data were categorized by the number of subjects reporting adverse events, body system, severity, seriousness, and relationship to test article. Comparisons among treatment groups were made by tabulating the frequency of subjects with one or more adverse events classified into MedDRA terms (Medical Dictionary for Regulatory Activities, http://www.meddramsso.com) during the study.
  • MedDRA terms Medical Dictionary for Regulatory Activities
  • Descriptive summary statistics for laboratory values and their associated change from baseline were determined for all clinical laboratory assessments. Values outside the normal range were flagged in the data listings.
  • shift tables were generated showing the number and percent of subjects that experienced changes in laboratory parameters during the course of the study (e.g., change from normal to high, based on the laboratory reference ranges).
  • Example 7 The following example describes a randomized, double blind, placebo-controlled study to evaluate the use of a hardy kiwifruit extract to decrease the CADESI score (Olivry et al, Vet. Dermatol, 13:77-87, 2002; Hanifm et al, Exp. Dermatol, 10:11-18, 2001; Kunz et al., Dermatology, 1997, 195, 10-19) of atopic dogs.
  • the objective of this study was to evaluate the efficacy of the A. arguta fruit extract FDOOl (PGl 02T) as an adjunct therapy to a standard steroid treatment for atopic dermatitis (AD) in dogs. Response to treatment was assessed using the investigator's global evaluation which incorporates the CADESI scale and the owner's Pruritis assessments. Additional objectives of the study were to assess the efficacy of the kiwifruit extract as a monotherapy to decrease the need for steroid use in the management of AD clinical signs, and to assess safety. Study design
  • a two week, low dose (0.2 mg/kg) steroid-only (prednisolone) period was administered to determine steroid responsiveness in the dogs, while retaining residual symptoms of AD. All dogs were healthy, excluding non-seasonal AD skin disease. Diagnosis of AD included at least three positive reactions to an intradermal skin allergy test. Dogs received flea control treatment (Advantage), and underwent a dietary trial to rule out food allergies as the main cause of symptomology. Dogs were free of concomitant medications such as antihistamines and long-acting injectable steroids, and free of any secondary infections. A minimum baseline (day 1) score of 25 on the CADESI scale was required for inclusion.
  • any dogs demonstrating improvement in their global evaluation will be advanced to a 28 day, open label second stage of the study, consisting of FDOOl test article (30 mg/kg/day) as a monotherapy for AD.
  • Subjects experiencing a relapse in AD signs, or requiring rescue medications will be considered Stage Two treatment failures and discontinued from the study.
  • Subjects maintaining their improvement in Stage Two will be considered treatment responders.
  • Assessment by the CADESI scale and Pruritis diaries will be done every seven days. Blood specimens will be collected during Stage Two on days 14 and 28. The laboratory blood panel will be identical to that of Stage One of the study.
  • the primary efficacy analysis will be the proportion of subjects with positive response to the treatment based on the investigator's global evaluation using the Chi-square test. An interim analysis may be conducted when the first half of the target enrollment completes the study. A p ⁇ 0.05 is considered significant.
  • Atopic dermatitis is the second most common allergy in dogs occurring in approximately 10% of canine population (Scott et al., Small Animal Dermatology, 5 th Ed., WB Saunders, 500-518, 1995).
  • AD in dogs tends to worsen with age.
  • Affected animals suffer from recurrent skin and ear infections that greatly decrease their quality of life.
  • available therapeutic options are limited.
  • Systemic treatments like glucocorticoids and cyclosporin may be effective but have the potential for adverse effects (Olivry et al., Vet. Dermatol, 13:77-87, 2002; Ryffel, et al., Arch. Toxicol., 53:107-141, 1983).
  • Glucocorticoids tend to be less effective with chronic use, oral cyclosporin may be cost-prohibitive in large breed dogs, and the success rate of antihistamines is often low (Scott et al., Small Animal Dermatology, 5 th Ed., WB Saunders, 500-518, 1995), 'Identification of a safe and effective treatment to decrease the signs and symptoms of canine AD would be of tremendous benefit.
  • Efficacy of FDOOl either as an adjunctive treatment or as a monotherapy, may decrease the need for steroid use in the management of AD in dogs. It is also envisioned that the kiwifruit extracts will be used as an adjunct therapy in order to decrease the potency of steroids used.
  • the extract could be administered as a capsule, a powder mixed with food, or as a component of the food. Dietary supplementation with kiwifruit preparations may be effective in supporting healthy skin in dogs with atopy when used alone or in combination with low dose steroids.
  • Example 8
  • the objective of this study will be to obtain preliminary evidence of effectiveness of FDOOl (PG102T), administered orally over a period of 28 days, in adult volunteers with allergic disease such as atopic dermatitis (AD), asthma, or allergic rhinitis of moderate severity.
  • a secondary objective of the study will be to measure the bioavailability of the product using alternative forms of delivery (e.g. capsules, a concentrate added to a beverage, emulsion, or food ingredient).
  • Another objective of the study will be to determine the effect of FDOOl on levels of pro-inflammatory blood markers such as cytokines, chemokines, leukotrienes, or antibodies, on the proliferation of myeloid cells (e.g. leukocytes, macrophages, mast cells, etc.), and on related gene expression and transcription factors.
  • a double-blind, placebo-controlled, outpatient study will be conducted in which subjects will be administered FDOOl as the test article or placebo.
  • Treatment arms of the study may include FDOOl as a monotherapy or as an adjunct therapy to oral steroids of mild to moderate potency.
  • steroid treatment for subjects with AD may be topical.
  • Steroid use may be as an intranasal spray or inhaler for asthma and allergic rhinitis subjects, respectively.
  • Subject inclusion criteria, efficacy assessments, safety, and tolerability, and statistical analyses will be assessed using methods similar to those described in Example 6.
  • One manifestation of allergy, atopic dermatitis is a common skin disorder in children and is usually observed during the first 6 months of life (Spergel and Paller, J. Allergy CHn.
  • AD Alzheimer's disease
  • kiwifruit extracts concentrates, other preparations, or extracts of other plant parts (e.g. bark, stem, roots, leaves), for adjunctive treatment or as a monotherapy for AD, asthma, allergic rhinitis, or other leukotriene-mediated conditions such as food allergies and chronic urticaria.
  • kiwifruit products as therapy for allergic conditions in small mammals (e.g. dogs, see also Example 7) may be explored further.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Pulmonology (AREA)
  • Oncology (AREA)
  • Medical Informatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Virology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Communicable Diseases (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)
  • Cosmetics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
PCT/US2006/006437 2005-02-25 2006-02-24 Compositions comprising actinidia and methods of use thereof WO2006093793A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
BRPI0608042-1A BRPI0608042A2 (pt) 2005-02-25 2006-02-24 composições compreendendo actinidia e métodos para o uso desta
MX2007010408A MX2007010408A (es) 2005-02-25 2006-02-24 Composiciones que comprenden actinidia y sus metodos de uso.
EP06735914A EP1858535A4 (en) 2005-02-25 2006-02-24 COMPOSITIONS WITH ACTINIDIA AND METHOD OF USE THEREOF
JP2007557161A JP2008531584A (ja) 2005-02-25 2006-02-24 マタタビ属を含む組成物およびそれを用いる方法
AU2006218875A AU2006218875B2 (en) 2005-02-25 2006-02-24 Compositions comprising Actinidia and methods of use thereof
US12/015,193 US20080175888A1 (en) 2005-02-25 2008-01-16 Combination Therapy Comprising Actinidia and Steroids and Uses Thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US65683905P 2005-02-25 2005-02-25
US65683805P 2005-02-25 2005-02-25
US60/656,839 2005-02-25
US60/656,838 2005-02-25

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11817216 A-371-Of-International 2006-02-24
US12/015,193 Continuation-In-Part US20080175888A1 (en) 2005-02-25 2008-01-16 Combination Therapy Comprising Actinidia and Steroids and Uses Thereof

Publications (3)

Publication Number Publication Date
WO2006093793A2 true WO2006093793A2 (en) 2006-09-08
WO2006093793A3 WO2006093793A3 (en) 2006-12-07
WO2006093793A8 WO2006093793A8 (en) 2008-03-27

Family

ID=36941647

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/006437 WO2006093793A2 (en) 2005-02-25 2006-02-24 Compositions comprising actinidia and methods of use thereof

Country Status (8)

Country Link
EP (1) EP1858535A4 (es)
JP (1) JP2008531584A (es)
AU (1) AU2006218875B2 (es)
BR (1) BRPI0608042A2 (es)
MX (1) MX2007010408A (es)
RU (1) RU2423139C2 (es)
TW (1) TWI385007B (es)
WO (1) WO2006093793A2 (es)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006055210A1 (de) * 2006-11-21 2008-05-29 Ionescu, John G., Dr. Diätisches Lebensmittel mit erhöhter freie Radikale bindender Wirkung
WO2009091915A1 (en) * 2008-01-16 2009-07-23 Efficas, Inc. Combination therapy comprising actinidia and steroids and uses thereof
KR101141182B1 (ko) 2009-05-12 2012-05-02 재단법인 제주테크노파크 항염증성 조성물
CN105410588A (zh) * 2015-11-06 2016-03-23 四川兴食尚科技有限公司 一种猕猴桃泡腾片及其制备工艺
US9352008B2 (en) 2009-08-12 2016-05-31 Melaleuca, Inc. Dietary supplements and methods for treating pain and inflammation
WO2019224200A1 (en) * 2018-05-22 2019-11-28 N.V. Nutricia Biomarkers for improving nutrion for infants at risk

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4750202B2 (ja) * 2009-10-16 2011-08-17 株式会社 藍匠 サルナシを用いた健康補助食品の製造方法
SG181975A1 (en) * 2009-12-29 2012-08-30 Nestec Sa Nutritional compositions comprising fruit flakes containing docosahexaenoic acid
JP5688233B2 (ja) * 2010-04-23 2015-03-25 ポーラ化成工業株式会社 Tarc産生抑制剤
JP6179840B2 (ja) * 2011-11-30 2017-08-16 国立大学法人 岡山大学 サルナシ由来の高極性有機溶媒抽出物とその利用
JP6242669B2 (ja) * 2013-11-27 2017-12-06 日本メナード化粧品株式会社 サルナシ抽出物を含有するヒアルロン酸生成促進剤

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5242814A (en) * 1989-10-10 1993-09-07 Eli Lilly And Company Polyether antibiotic
DE19758090B4 (de) * 1997-12-18 2008-10-16 Maria Endres Zusammensetzung und deren Verwendung in Kosmetik und Medizin
WO2002079372A1 (en) * 2001-03-29 2002-10-10 Council Of Scientific And Industrial Research Process for the preparation of herbal wines from himalayan berriers
US6811796B2 (en) * 2002-04-22 2004-11-02 Matsuura Yakugyo Co., Ltd. Preventive or therapeutic agent for pollen allergy, allergic rhinitis, atopic dermatitis, asthma or urticaria, or health food for prevention or improvement or reduction of symptoms thereof
WO2003103415A1 (en) * 2002-06-07 2003-12-18 Ann De Wees Allen Novel sweetener compositions and methods of use
KR100615389B1 (ko) * 2002-08-23 2006-08-25 (주)헬릭서 다래 추출물을 함유하는 알러지성 질환 및 비알러지성염증 질환의 예방 및 개선용 건강 기능 식품

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None
See also references of EP1858535A4

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006055210A1 (de) * 2006-11-21 2008-05-29 Ionescu, John G., Dr. Diätisches Lebensmittel mit erhöhter freie Radikale bindender Wirkung
WO2009091915A1 (en) * 2008-01-16 2009-07-23 Efficas, Inc. Combination therapy comprising actinidia and steroids and uses thereof
KR101141182B1 (ko) 2009-05-12 2012-05-02 재단법인 제주테크노파크 항염증성 조성물
US9352008B2 (en) 2009-08-12 2016-05-31 Melaleuca, Inc. Dietary supplements and methods for treating pain and inflammation
US10653734B2 (en) 2009-08-12 2020-05-19 Melaleuca, Inc. Dietary supplements and methods for treating pain and inflammation
CN105410588A (zh) * 2015-11-06 2016-03-23 四川兴食尚科技有限公司 一种猕猴桃泡腾片及其制备工艺
WO2019224200A1 (en) * 2018-05-22 2019-11-28 N.V. Nutricia Biomarkers for improving nutrion for infants at risk

Also Published As

Publication number Publication date
WO2006093793A3 (en) 2006-12-07
RU2423139C2 (ru) 2011-07-10
TW200716226A (en) 2007-05-01
TWI385007B (zh) 2013-02-11
EP1858535A2 (en) 2007-11-28
JP2008531584A (ja) 2008-08-14
BRPI0608042A2 (pt) 2009-11-03
AU2006218875A1 (en) 2006-09-08
RU2007135365A (ru) 2009-03-27
EP1858535A4 (en) 2012-05-02
AU2006218875B2 (en) 2012-08-09
MX2007010408A (es) 2008-03-25
WO2006093793A8 (en) 2008-03-27

Similar Documents

Publication Publication Date Title
AU2006218875B2 (en) Compositions comprising Actinidia and methods of use thereof
US20100111927A1 (en) Compositions Comprising Actinidia and Methods of Use Thereof
EP2329835B1 (en) Composition comprising the extract of actinidia arguta and related species for the prevention and treatment of allergic disease and non-allergic inflammatory disease
US20080175888A1 (en) Combination Therapy Comprising Actinidia and Steroids and Uses Thereof
JP7277571B2 (ja) イノトジオール化合物を有効成分として含む、アレルギー疾患の予防又は治療用組成物
CN101291679A (zh) 包含猕猴桃属植物的组合物及其使用方法
NZ587187A (en) Compositions Comprising Actinidia ( Kiwifruit) and a Steriod Methods of Use Thereof in Manufacture of Medicament
KR101150485B1 (ko) 알코니아 트리플너비아 추출물을 함유하는 천식 또는 염증성 질환의 예방 및 치료용 약학적 조성물
KR101349747B1 (ko) 워크리아 인시그니스 추출물 또는 이의 분획물을 함유하는 염증성 질환 또는 천식의 예방 및 치료용 조성물
KR101333715B1 (ko) 마스티시아 알보리아 추출물 또는 이의 분획물을 유효성분으로 함유하는 염증성 질환 예방 및 치료용 약학적 조성물
KR20200094480A (ko) 나도하수오 추출물을 이용한 항알러지 개선용 조성물
KR20200021739A (ko) 분꽃나무 추출물을 포함하는 호흡기 질환 예방, 개선 또는 치료용 조성물
KR20180075241A (ko) 아토피성 피부질환 예방 또는 치료용 조성물

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680013923.4

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/010408

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2007557161

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 561749

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2006218875

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006735914

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 7367/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007135365

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2006218875

Country of ref document: AU

Date of ref document: 20060224

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0608042

Country of ref document: BR

Kind code of ref document: A2