CA2703698A1 - Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent - Google Patents

Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent Download PDF

Info

Publication number
CA2703698A1
CA2703698A1 CA2703698A CA2703698A CA2703698A1 CA 2703698 A1 CA2703698 A1 CA 2703698A1 CA 2703698 A CA2703698 A CA 2703698A CA 2703698 A CA2703698 A CA 2703698A CA 2703698 A1 CA2703698 A1 CA 2703698A1
Authority
CA
Canada
Prior art keywords
cox
free
flavans
composition
acacia
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA2703698A
Other languages
French (fr)
Other versions
CA2703698C (en
Inventor
Qi Jia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Unigen Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2703698A1 publication Critical patent/CA2703698A1/en
Application granted granted Critical
Publication of CA2703698C publication Critical patent/CA2703698C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

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)
    • A61K36/53Lamiaceae or Labiatae (Mint family), e.g. thyme, rosemary or lavender
    • A61K36/539Scutellaria (skullcap)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • 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
    • A61K36/11Pteridophyta or Filicophyta (ferns)
    • 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
    • A61K36/13Coniferophyta (gymnosperms)
    • A61K36/15Pinaceae (Pine family), e.g. pine or cedar
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/28Asteraceae or Compositae (Aster or Sunflower family), e.g. chamomile, feverfew, yarrow or echinacea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/47Euphorbiaceae (Spurge family), e.g. Ricinus (castorbean)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/48Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/48Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
    • A61K36/484Glycyrrhiza (licorice)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/48Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
    • A61K36/486Millettia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/53Lamiaceae or Labiatae (Mint family), e.g. thyme, rosemary or lavender
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/54Lauraceae (Laurel family), e.g. cinnamon or sassafras
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/60Moraceae (Mulberry family), e.g. breadfruit or fig
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/74Rubiaceae (Madder family)
    • 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)
    • A61K36/88Liliopsida (monocotyledons)
    • A61K36/906Zingiberaceae (Ginger family)
    • 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)
    • A61K36/88Liliopsida (monocotyledons)
    • A61K36/906Zingiberaceae (Ginger family)
    • A61K36/9062Alpinia, e.g. red ginger or galangal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • 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
    • 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
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Abstract

The present invention provides a novel composition of matter comprised of a mixture of two specific classes of compounds --Free-B-ring flavonoids and flavans-- for use in the prevention and treatment of diseases and conditions mediated by the COX-2 and 5-LO pathways. The present invention further provides a novel method for simultaneously inhibiting the cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO) enzymes, and reducing cox-2 mRNA production. Finally, the present invention includes a method for weight loss and blood glucose control. The methods of this invention are comprised of administering to a host in need thereof an effective amount of the composition of this invention together with a pharmaceutically acceptable carrier.; This invention relates generally to the prevention and treatment of diseases and conditions mediated by the cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO) pathways, including but not limited to the relief joint discomfort and pain associated with conditions such as osteoarthritis, rheumatoid arthritis, and other injuries that result from overuse.

Description

FORMULATION OF A MIXTURE OF FREE-B-RING FLAVONOIDS AND
FLAVANS AS A THERAPEUTIC AGENT

FIELD OF THE INVENTION
This invention relates generally to the prevention and treatment of diseases and conditions mediated by the cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO) pathways. Specifically, the present invention relates to a novel composition of matter comprised of a mixture of a blend of two specific classes of compounds --Free-B-ring flavonoids and flavans-- for use in the prevention and treatment of diseases and conditions mediated by the COX-2 and 5-LO pathways. Included in the present invention is a method for the simultaneous inhibition of the protein function of the COX-2 and 5-LO
enzymes, and a method for modulating the production of mRNA by administration of the novel composition of this invention. Also included in the present invention is a method for the prevention and treatment of COX-2 and 5-LO mediated diseases and conditions, including but not limited to joint discomfort and pain associated with conditions such as osteoarthritis, rheumatoid arthritis, and other injuries that result from overuse. Further included in the present invention is a method for reducing blood glucose levels and promoting weight loss.

BACKGROUND OF THE INVENTION
The liberation and metabolism of arachidonic acid (AA) from the cell membrane results in the generation of pro-inflammatory metabolites by several different pathways.
Arguably, two of the most important pathways to inflammation are mediated by the enzymes 5-lipoxygenase (5-LO) and cyclooxygenase (COX). These parallel pathways result in the generation of leukotrienes and prostaglandin, respectively, which play important roles in the initiation and progression of the inflammatory response. These vasoactive compounds are chemotaxins, which promote infiltration of inflammatory cells into tissues and serve to prolong the inflammatory response. Consequently, the enzymes responsible for generating these mediators of inflammation have become the targets for many new drugs aimed at the treatment of inflammation that contributes to the pathogenesis of diseases such as rheumatoid arthritis, osteoarthritis, Alzheimer's disease and certain types of cancer.
Inhibition of the cyclooxygenase (COX) enzyme is the mechanism of action attributed to most nonsteroidal anti-inflammatory drugs (NSAIDS). There are two distinct isoforms of the COX enzyme (COX-1 and COX-2) that share approximately 60%

sequence homology, but differ in expression profiles and function. COX-1 is a constitutive form of the enzyme that has been linked to the production of physiologically important prostaglandin involved in the regulation of normal physiological functions such as platelet aggregation, protection of cell function in the stomach and maintenance of normal kidney function (Dannhardt and Kiefer (2001) Eur. J. Med. Chem. 36:109-26).
The second isoform, COX-2, is a form of the enzyme that is inducible by pro-inflammatory cytokines such as interleukin-1(3 (IL-1(3) and other growth factors (Herschmann (1994) Cancer Metastasis Rev. 134:241-56; Xie et al. (1992) Drugs Dev.
Res. 25:249-65). This isoform catalyzes the production of prostaglandin E2 (PGE2) from AA. Inhibition of COX-2 is responsible for the anti-inflammatory activities of conventional NSAIDs.
Inhibitors that demonstrate dual specificity for COX-2 and 5-LO, while maintaining COX-2 selectivity relative to COX-1, would have the obvious benefit of inhibiting multiple pathways of AA metabolism. Such inhibitors would block the inflammatory effects of PGE2, as well as, those of multiple leukotrienes (LT) by limiting their production. This includes the vasodilation, vasopermeability and chemotactic effects of LTB4 and LTD4 and the effects of LTE4, also known as the slow reacting substance of anaphalaxis. Of these, LTB4 has the most potent chemotactic and chemokinetic effects (Moore (1985) in Prostanoids: Pharmacological, Physiological and Clinical Relevance Cambridge University Press, N.Y., pp. 229-30) and has been shown to be elevated in the gastrointestinal mucosa of patients with inflammatory bowel disease (Sharon and Stenson (1983) Gastroenterology 84:1306-13) and within the synovial fluid of patients with rheumatoid arthritis (Klicksein et al. (1980) J. Clin. Invest. 66:1166-70; Rae et al. (1982) Lancet ii:1122-4).

In addition to the above-mentioned benefits of dual COX-2/5-LO inhibitors, many dual inhibitors do not cause some of the side effects that are typical of NSAIDs or COX-2 inhibitors, including the gastrointestinal damage and discomfort caused by traditional NSAIDs. It has been suggested that NSAID-induced gastric inflammation is largely due to metabolites of 5-LO, particularly LTB4, which attracts cells to the site of a gastric lesion thus causing further damage (Kircher et al. (1997) Prostaglandins Leukot.
Essent. Fatty Acids 56:417-23). Leukotrienes represent the primary AA metabolites within the gastric mucosa following prostanoid inhibition. It appears that these compounds contribute to a significant amount of the gastric epithelial injury resulting from the use of NSAIDs.
(Celotti and Laufer (2001) Pharmacol. Res. 43:429-36). Dual inhibitors of COX-2 and 5-LO were also demonstrated to inhibit the coronary vasoconstriction in arthritic hearts in a rat model (Gok et al. (2000) Pharmacology 60:41-46). Taken together, these characteristics suggest that there may be distinct advantages to dual inhibitors of COX-2 and 5-LO over specific COX-2 inhibitors and non-specific NSAIDs with regard to both increased efficacy and reduced side effects.

Because the mechanism of action of COX inhibitors overlaps that of most conventional NSAIDs, COX inhibitors are used to treat many of the same symptoms, such as the pain and swelling associated with inflammation in transient conditions and chronic diseases in which inflammation plays a critical role. Transient conditions include the treatment of inflammation associated with minor abrasions, sunburn or contact dermatitis, as well as, the relief of pain associated with tension and migraine headaches and menstrual cramps. Chronic conditions include arthritic diseases such as rheumatoid arthritis and osteoarthritis. Although rheumatoid arthritis is largely an autoinirnune disease and osteoarthritis is caused by the degradation of cartilage in joints, reducing the inflammation associated with each provides a significant increase in the quality of life for those suffering from these diseases (Wienberg (2001) Immunol. Res. 22:319-41; Wollhiem (2000) Curr.
Opin. Rheum. 13:193-201). As inflammation is a component of rheumatic diseases in general, the use of COX inhibitors has been expanded to include diseases such as systemic lupus erythromatosus (SLE) (Goebel et al. (1999) Chem. Res. Tox. 12:488-500;
Patrono et al. (1985) J. Clin. Invest. 76:1011-1018) and rheumatic skin conditions such as scleroderma. COX inhibitors are also used for the relief of inflammatory skin conditions that are not of rheumatic origin, such as psoriasis, in which reducing the inflammation resulting from the over production of prostaglandins could provide a direct benefit (Fogh et al. (1993) Acta Derm. Venereol (Oslo) 73:191-3).
In addition to their use as anti-inflammatory agents, another potential role for COX
inhibitors is the treatment of cancer. Over-expression of COX-2 has been demonstrated in various human malignancies and inhibitors of COX-2 have been shown to be efficacious in the treatment of animals with skin, breast and bladder tumors. While the mechanism of action is not completely understood, the over-expression of COX-2 has been shown to inhibit apoptosis and increase the invasiveness of tumorgenic cell types (Dempke et al.
(2001) J. Can. Res. Clin. Oncol. 127:411-17; Moore and Simmons (2000) Current Med.

Chem. 7:1131-44). It is possible that enhanced production of prostaglandins, resulting from the over-expression of COX-2, promotes cellular proliferation and consequently increases angiogenesis. (Moore (1985) in Prostanoids: Pharmacological, Physiological and Clinical Relevance, Cambridge University Press, N.Y., pp. 229-30; Fenton et al.
(2001) Am. J. Clin. Oncol. 24:453-57).
There have been a number of clinical studies evaluating COX-2 inhibitors for potential use in the prevention and treatment of different types of cancer. In 1999, 130,000 new cases of colorectal cancer were diagnosed in the United States. Aspirin, a non-specific NSAID, has been found to reduce the incidence of colorectal cancer by 40-50%
(Giovannucci et al. (1995) N. Engl. J. Med. 333:609-614) and mortality by 50%
(Smalley et al. (1999) Arch. Intern. Med. 159:161-166). In 1999, the FDA approved the inhibitor celecoxib for use in FAP (Familial Ademonatous Polyposis) to reduce colorectal cancer mortality. It is believed that other cancers with evidence of COX-2 involvement may be successfully prevented and/or treated with COX-2 inhibitors including, but not limited to, esophageal cancer, head and neck cancer, breast cancer, bladder cancer, cervical cancer, prostate cancer, hepatocellular carcinoma and non-small cell lung cancer (Jaeckel et al. (2001) Arch. Otolarnygol. 127:1253-59; Kirschenbaum et al. (2001) Urology 58:127-31; Dannhardt and Kiefer (2001) Eur. J. Med. Chem. 36:109-26). COX-2 inhibitors may also prove 'successful in preventing colon cancer in high-risk patients.
There is also evidence that COX-2 inhibitors can prevent or even reverse several types of life-threatening cancers. To date, as many as fifty studies show that COX-2 inhibitors can prevent pre-malignant and malignant tumors in animals, and possibly prevent bladder, esophageal and skin cancers as well. COX-2 inhibition could prove to be one of the most important preventive medical accomplishments of the century.
Recent scientific progress has identified correlations between COX-2 expression, general inflammation and the pathogenesis of Alzheimer's Disease (AD) (Ho et at. (2001) 5 Arch. Neurol. 58:487-92). In animal models, transgenic mice that over-express the COX-2 enzyme have neurons that are more susceptible to damage. The National Institute on Aging (NIA) is launching a clinical trial to determine whether NSAIDs can slow the progression of Alzheimer's disease. Naproxen (a non-selective NSAID) and rofecoxib (Vioxx, a COX-2 specific selective NSAID) will be evaluated. Previous evidence has indicated that inflammation contributes to Alzheimer's disease. According to the Alzheimer's Association and the NIA, about 4 million people suffer from AD in the United States and this is expected to increase to 14 million by mid-century.
The COX enzyme (also known as prostaglandin H2 synthase) catalyzes two separate reactions. In the first reaction, AA is metabolized to form the unstable prostaglandin G2 (PGG2), a cyclooxygenase reaction. In the second reaction, PGG2 is converted to the endoperoxide PGH2, a peroxidase reaction. The short-lived PGH2 non-enzymatically degrades to PGE2. The compounds described herein are the result of a discovery strategy that combined an assay focused on the inhibition of COX-1 and COX-2 peroxidase activity with a chemical dereplication process to identify novel inhibitors of the COX enzymes.
The term gene expression is often used to describe the broad result of mRNA
production and protein synthesis. In fact, changes in actual gene expression may never result in observable changes on the protein level. The corollary, that changes in protein level do not always result from changes in gene expression, can also be true.
There are six possible points of regulation in the pathway leading from genomic DNA to a functional protein: (1) transcriptional regulation by nuclear factors and other signals leading to production of pre-mRNA; (2) pre-mRNA processing regulation involving exon splicing, the additions of a 5' cap structure and 3' poly-adenylation sequence and transport of the mature mRNA from the nucleus into the cytoplasm; (3) mRNA transport regulation controlling localization of the mRNA to a specific cytoplasmic site for translation into protein; (4) mRNA degradation regulation controlling the size of the mRNA pool either prior to any protein translation or as a means of ending translation from that specific mRNA; (5) translational regulation of the specific rate of protein translation initiation and (6) post-translation processing regulation involving modifications such as glycosylation and proteolytic cleavage. In the context of genomics research it is important to use techniques that measure gene expression levels closer to the initial steps (e.g. mRNA
levels) rather than later steps (e.g. protein levels) in this pathway.
Recent reports have addressed the possible involvement of flavonoids, isolated from the medicinal herb Scutellaria baicalensis, in alterations in cox-2 gene expression (Wakabayashi and Yasui (2000) Eur. J. Pharmacol. 406:477-481; Chen et al.
(2001) Biochem. Pharmacol. 61:1417-1427; Chi et al. (2001) 61:1195-1203 and Raso et al.
(2001) Life Sci. 68:921-931). Each of above cited studies on cox-2 gene expression used a Western Blot technique to evaluate putative alterations in gene expression without validation on the molecular level. Since this method only measures protein levels and not the specific transcription product, mRNA, it is possible that other mechanisms are involved leading to the observed increase in protein expression. For example, LPS has been reported to modulate mRNA half-lives via instability sequences found in the 3' untranslated region (3'UTR) of mRNAs (Watkins et al. (1999) Life Sci. 65:449-481), which could account for increased protein expression without alternations in the rate of gene transcription. Consequently, this leaves open the question of whether or not these treatment conditions resulted in a meaningful change in gene expression.

Techniques, such as RT-qPCR and DNA microarray analysis, rely on mRNA levels for analysis and can be used to evaluate levels of gene expression under different conditions, i.e. in the presence or absence of a pharmaceutical agent. There are no known reports using techniques that specifically measure the amount of mRNA, directly or indirectly, in the literature when Free-B-ring flavonoids or flavans are used as the therapeutic agents.
Flavonoids are a widely distributed group of natural products. The intake of flavonoids has been demonstrated to be inversely related to the risk of incident dementia.
The mechanism of action, while not known, has been speculated as being due to the anti-oxidative effects of flavonoids (Commenges et al. (2000) Eur. J. Epidemiol.
16:357-363).
Polyphenol flavones induce programmed cell death, differentiation and growth inhibition in transformed colonocytes by acting at the mRNA level on genes including cox-2, Nuclear Factor kappa B (NFiB) and bcl-X(L) (Wenzel et al. (2000) Cancer Res.
60:3823-3831). It has been reported that the number of hydroxyl groups on the B ring is important in the suppression of cox-2 transcriptional activity (Mutoh et al. (2000) Jnp.
J. Cancer Res.
91:686-691).

Free-B-ring flavones and flavonols are a specific class of flavonoids, which have no substituent groups on the aromatic B ring (referred to herein as Free-B-ring flavonoids), as illustrated by the following general structure:

R2 Rs A C

wherein R1, R2, R3, R4, and R5 are independently selected from the group consisting of -H, -OH, -SH, OR, -SR, -NH2, -NHR, -NR2, -NR3{X , a carbon, oxygen, nitrogen or sulfur, glycoside of a single or a combination of multiple sugars including, but not limited to aldopentoses, methyl-aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof;
wherein R is an alkyl group having between 1-10 carbon atoms; and X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, carbonate, etc.

Free-B-ring flavonoids are relatively rare. Out of 9,396 flavonoids synthesized or isolated from natural sources, only 231 Free-B-ring flavonoids are known (The Combined Chemical Dictionary, Chapman & Hall/CRC, Version 5:1 June 2001). Free-B-ring flavonoids have been reported to have diverse biological activity. For example, galangin (3,5,7-trihydroxyflavone) acts as an anti-oxidant and free radical_ scavenger and is believed to be a promising candidate for anti-genotoxicity and cancer chemoprevention.
(Heo et al.
(2001) Mutat. Res. 488:135-150). It is an inhibitor of tyrosinase monophenolase (Kubo et al. (2000) Bioorg. Med. Chem. 8:1749-1755), an inhibitor of rabbit heart carbonyl reductase (Imamura et al. (2000) J. Biochem. 127:653-658), has antimicrobial activity (Afolayan and Meyer (1997) Ethnopharmacol. 57:177-181) and antiviral activity (Meyer et al. (1997) J. Ethnopharmacol. 56:165-169). Baicalein and two other Free-B-ring flavonoids, have antiproliferative activity against human breast cancer cells.
(So et al.
(1997) Cancer Lett. 112:127-133).
Typically, flavonoids have been tested for activity randomly based upon their availability. Occasionally, the requirement of substitution on the B-ring has been emphasized for specific biological activity, such as the B-ring substitution required for high affinity binding to p-glycoprotein (Boumendjel et al. (2001) Bioorg. Med.
Chem.
Lett. 11:75-77); cardiotonic effect (Itoigawa et al. (1999) J. Ethnopharmacol.
65: 267-272), protective effect on endothelial cells against linoleic acid hydroperoxide-induced toxicity (Kaneko and Baba (1999) Biosci. Biotechnol. Biochem. 63:323-328), COX-inhibitory activity (Wang (2000) Phytomedicine 7:15-19) and prostaglandin endoperoxide synthase activity (Kalkbrenner et al. (1992) Pharmacology 44:1-12). Only a few publications have mentioned the significance of the unsubstituted B-ring of the Free-B-ring flavonoids. One example is the use of 2-phenyl flavones, which inhibit NADPH
quinone acceptor oxidoreductase, as potential anticoagulants (Chen et al.
(2001) Biochem.
Pharmacol. 61:1417-1427).
The reported mechanism of action with respect to the anti-inflammatory activity of various Free-B-ring flavonoids has been controversial. The anti-inflammatory activity of the Free-B-ring flavonoids, chrysin (Liang et al. (2001) FEBS Lett. 496:12-18), wogonin (Chi et al. (2001) Biochem. Pharmacol. 61:1195-1203) and halangin (Raso et al.
(2001) Life Sci. 68:921-931) has been associated with the suppression of inducible cyclooxygenase and nitric oxide synthase via activation of peroxisome-proliferator activated receptor gamma (PPARy) and influence on degranulation and AA release (Tordera et al. (1994) Z. Naturforsch [C] 49:235-240). It has been reported that oroxylin, baicalein and wogonin inhibit 12-lipoxygenase activity without affecting cyclooxygenases (You et al. (1999) Arch. Pharm. Res. 22:18-24). More recently, the anti-inflammatory activity of wogonin, baicalin and baicalein has been reported as occurring through inhibition of inducible nitric oxide synthase and cox-2 enzyme production induced by nitric oxide inhibitors and lipopolysaccharides (Chen et al. (2001) Biochem.
Pharmacol.
61:1417-1427). It has also been reported that oroxylin acts via suppression of NFicB
activation (Chen et al. (2001) Biochem. Pharmacol. 61:1417-1427). Finally, wogonin reportedly inhibits inducible PGE2 production in macrophages (Wakabayashi and Yasui (2000)' Eur. J. Pharmacol. 406:477-481).
Inhibition of the phosphorylation of mitogen-activated protein kinase (MAPK) and inhibition of Ca2+ ionophore A23187 induced PGE2 release by baicalein has been reported as the mechanism of anti-inflammatory activity of Scutellariae radix (Nakahata et al.

(1999) Nippon Yakurigalcu Zasshi 114, Supp.11:215P-219P; Nakahata et al.
(1998) Am. J.
Chin. Med. 26:311-323). Baicalin from Scutellaria baicalensis reportedly inhibits superantigenic staphylococcal exotoxins stimulated T-cell proliferation and production of IL-lei, IL-6, tumor necrosis factor-a (TNF-a), and interferon-7 (IFN-y) (Krakauer et al.
(2001) FEBS Lett. 500:52-55). Thus, the anti-inflammatory activity of baicalin has been associated with inhibiting the pro-inflammatory cytokines mediated signaling pathways activated by superantigens. However, it has also been proposed that the anti-inflammatory activity of baicalin is due to the binding of a variety of chemokines, which limit their biological activity (Li et al. (2000) Immunopharmacol. 49:295-306). Recently, the effects of baicalin on adhesion molecule expression induced by thrombin and thrombin receptor agonist peptide (Kimura et al. (2001) Planta Med. 67:331-334), as well as, the inhibition of MAPK cascade (Nakahata et al. (1999) Nippon Yakurigaku Zasshi 114, Supp 11:215P-219P; Nakahata et al. (1998) Am. J. Chin Med. 26:311-323) have been reported.
The Chinese medicinal plant Scutellaria baicalensis contains significant amounts of Free-B-ring flavonoids, including baicalein, baicalin, wogonin * and baicalenoside.
Traditionally, this plant has been used to treat a number of conditions including clearing away heat, purging fire, dampness-warm and summer fever syndromes; polydipsia resulting from high fever; carbuncle, sores and other pyogenic skin infections; upper respiratory infections such as acute tonsillitis, laryngopharyngitis and scarlet fever; viral hepatitis; nephritis; pelvitis; dysentery; hematemesis and epistaxis. This plant has also traditionally been used to prevent miscarriage (see Encyclopedia of Chinese Traditional Medicine, ShangHai Science and Technology Press, ShangHai, China, 1998).
Clinically, Scutellaria is now used to treat conditions such as pediatric pneumonia, pediatric bacterial diarrhea, viral hepatitis, acute gallbladder inflammation, hypertension, topical acute inflammation resulting from cuts and surgery, bronchial asthma and upper respiratory infections (Encyclopedia of Chinese Traditional Medicine, ShangHai Science and 5 Technology Press, ShangHai, China, 1998). The pharmacological efficacy of Scutellaria roots for treating bronchial asthma is reportedly related to the presence of Free-B-ring flavonoids and their suppression of eotaxin associated recruitment of eosinophils (Nakajima et al. (2001) Planta Med. 67(2):132-135).
To date, a number of naturally occurring Free-B-ring flavonoids have been 10 commercialized for varying uses. For example, liposome formulations of Scutellaria extracts have been utilized for skin care (U.S. Pat. Nos. 5,643,598;
5,443,983). Baicalin has been used for preventing cancer due to its inhibitory effects on oncogenes (U.S. Pat.
No. 6,290,995). Baicalin and other compounds have been used as antiviral, antibacterial and immunomodulating agents (U.S. Pat. No. 6,083,921) and as natural anti-oxidants (Poland Pub. No. 9,849,256). Chrysin has been used for its anxiety reducing properties (U.S. Pat. No. 5,756,538). Anti-inflammatory flavonoids are used for the control and treatment of anorectal and colonic diseases (U.S. Pat. No. 5,858,371) and inhibition of lipoxygenase (U.S. Pat. No. 6,217,875). These compounds are also formulated with glucosamine collagen and other ingredients for repair and maintenance of connective tissue (U.S. Pat. No. 6,333,304). Flavonoid esters constitute the active ingredients for cosmetic compositions (U.S. Pat. No. 6,235,294). U.S. Application Serial No.
10/091,362, filed March 1, 2002, entitled "Identification of Free-B-ring Flavonoids as Potent COX-2 Inhibitors," discloses a method for inhibiting the cyclooxygenase enzyme COX-2 by administering a composition comprising a Free-B-ring flavonoid or a composition containing a mixture of Free-B-ring flavonoids to a host in need thereof. This is the first report of a link between Free-B-ring flavonoids and COX-2 inhibitory activity.
This application is specifically incorporated herein by reference in its entirety.
Japanese Pat. No. 63027435, describes the extraction, and enrichment of baicalein and Japanese Pat. No. 61050921 describes the purification of baicalin.
Flavans include compounds illustrated by the following general structure:
wherein R1, R2, R3, R4 and R5 are independently selected from the group consisting of -H, -OH, -SH, -OCH3, -SCH3, -OR, -SR, -NH2, -NRH, -NR2, -NR3+X-, esters of the mentioned J substitution groups, including, but not limited to, gallate, acetate, cinnamoyl and hydroxyl-cinnamoyl esters, trihydroxybenzoyl esters and caffeoyl esters, and their chemical derivatives thereof; a carbon, oxygen, nitrogen or sulfur glycoside of a single or a combination of multiple sugars including, but not limited to, aldopentoses, methyl aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof;
dimer, trimer and other polymerized flavans;
wherein R is an alkyl group having between 1-10 carbon atoms; and X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, and carbonate, etc.

Catechin is a flavan, found primarily in Acacia, having the following structure OH
OH
HO o \ o x OH
Catechin Catechin works both alone and in conjunction with other flavonoids found in tea, and has both antiviral and antioxidant activity. Catechin has been shown to be effective in the treatment of viral hepatitis. It also appears to prevent oxidative damage to the heart, kidney, lungs and spleen and has been shown to inhibit the growth of stomach cancer cells.
Catechin and its isomer epicatechin inhibit prostaglandin endoperoxide synthase with an IC50 value of 40 M. (Kalkbrenner et al. (1992) Pharmacol. 44:1-12).
Five flavan-3-ol derivatives, including (+)-catechin and gallocatechin, isolated from the four plant species, Atuna racemosa, Syzygium caiynocarpum, Syzygiunz malaccense and Vantanea peruviana, exhibit equal to or weaker inhibitory activity against COX-2, relative to COX-1, with IC50 values ranging from 3.3 M to 138 M (Noreen et al. (1998) Planta Med. 64:520-524). (+)-Catechin, isolated from the bark of Ceiba pentandra, inhibits COX-1 with an IC50 value of 80 M (Noreen et al. (1998) J. Nat. Prod. 61:8-12).
Commercially available pure (+)-catechin inhibits COX-1 with an IC50 value of around 183 to 279 M, depending upon the experimental conditions, with no selectivity for COX-2 (Noreen et al. (1998) J. Nat. Prod. 61:1-7).
Green tea catechin, when supplemented into the diets of Sprague dawley male rats, lowered the activity level of platelet PLA2 and significantly reduced platelet cyclooxygenase levels (Yang et al. (1999) J. Nutr. Sci. Vitaminol. 45:337-346). Catechin and epicatechin reportedly weakly suppress cox-2 gene transcription in human colon cancer DLD-1 cells (IC50 = 415.3 AM) (Mutoh et al. (2000) Jpn. J. Cancer Res.
91:686-691). The neuroprotective ability of (+)-catechin from red wine results from the antioxidant properties of catechin, rather than inhibitory effects on intracellular enzymes, such as cyclooxygenase, lipoxygenase or nitric oxide synthase (Bastianetto et al. (2000) Br. J. Pharmacol. 131:711-720). Catechin derivatives purified from green tea and black tea, such as epigallocatechin-3-gallate (EGCG), epigallocatechin (EGC), epicatechin-3-gallate (ECG) and theaflavins showed inhibition of cyclooxygenase- and lipoxygenase-dependent metabolism of AA in human colon mucosa and colon tumor tissues (Hong et al.
(2001) Biochem. Pharmacol. 62:1175-1183) and induced cox-2 gene expression and production (Park et al. (2001) Biochem. Biophys. Res. Con-nnun. 286:721-725).
Epiafzelechin isolated from the aerial parts of Celastrus orbiculatus exhibited dose-dependent inhibition of COX-1 activity with an IC50 value of 15 M and also demonstrated anti-inflammatory activity against carrageenin-induced mouse paw edema following oral administration at a dosage of 100 mg/kg (Min et al. (1999) Planta Med.
65:460-462).
Catechin and its derivatives from various plant sources, especially from green tea leaves, have been used in the treatment of HPV infected Condyloma acuminata (Cheng, U.S. Pat. No. 5,795,911) and in the treatment of hyperplasia caused by papilloma virus (Cheng, U.S. Pat. Nos. 5,968,973 and 6,197,808). Catechin and its derivatives have also been used topically to inhibit angiogenesis in mammalian tissue, in conditions such as skin cancer, psoriasis, spider veins or under eye circles (Anderson, U.S. Pat. No.
6,248,341), against UVB-induced tumorigenesis in mice (Agarwal et al. (1993) Photochem.
Photobiol.
58:695-700), for inhibiting nitric oxide synthase at the level of gene expression and enzyme activity (Chan, U.S. Pat. No. 5,922,756), and as hair-growing agents (Takahashi, U.S. Pat. No. 6,126,940). Catechin-based compositions have also been formulated with other extracts and vitamins for treatment of acne (Murad, U.S. Pat. No.
5,962,517), hardening the tissue of digestive organs (Shi, U.S. Pat. No. 5,470, 589) and for inhibiting 5 alpha-reductase activity in treating androgenic disorder related diseases and cancers (Liao, U.S. Pat. No. 5,605,929). Green tea extract has been formulated with seven other plant extracts for reducing inflammation by inhibiting the COX-2 enzyme, without identification of any of the specific active components (Mewmark, U.S. Pat. No. 6,264,995).
Acacia is a genus of leguminous trees and shrubs. The genus Acacia includes more than 1,000 species belonging to the family of Leguminosae and the subfamily of Mimosoideae. Acacias are distributed worldwide in places such as tropical and subtropical areas of Central and South America, Africa, parts of Asia, as well as, Australia, which has the largest number of endemic species. Acacias are present primarily in dry and and regions where the forests are often in the nature of open thorny shrubs.
The genus Acacia is divided into 3 subgenera based mainly on leaf morphology --Acacia, Aculiferum and Heterophyllum. Based on the nature of the leaves of mature trees, however, the genus Acacia can be divided into two "popular" groups --the typical bipinnate-leaved species and the phyllodenous species. A phyllode is a modified petiole expanded into a leaf-like structure with no leaflets, an adaptation to xerophytic conditions. The typical bipinnate-leaved species are found primarily throughout the tropics, whereas the phyllodenous species occur mainly in Australia. More than 40 species of Acacia have been reported in India. Gamble in his book entitled Flora of Madras Presidency listed 23 native species for southern India, 15 of which are found in Tamil Nadu. Since that time, however, many new Acacia species have been introduced to India and approximately 40 species are now found in Tamil Nadu itself The indigenous species are primarily thorny trees or shrubs and a few are thorny stragglers, such as A. caesia, A. pennata and A. sinuata. Many species have been introduced from Africa and Australia, including A. mearnsii, A. picnantha and A.

dealbata, which have bipinnate leaves and A. auriculiformis, A. holoserecia and A.
mangium, which are phyllodenous species.

Acacias are very important economically, providing a source of tannins, gums, timber, fuel and fodder. Tannins, which are isolated primarily from the bark, are used extensively for tanning hides and skins. Some Acacia barks are also used for flavoring local spirits. Some indigenous species like A. sinuata also yield saponins, which are any of various plant glucosides that form soapy lathers when mixed and agitated with water.
Saponins are used in detergents, foaming agents and emulsifiers. The flowers of some Acacia species are fragrant and used to make perfume. For example, cassie perfume is obtained from A. ferrugenea. The heartwood of many Acacias is used for making agricultural implements and also provides a source of firewood. Acacia gums find extensive use in medicine and confectionary and as sizing and finishing materials in the textile industry. Lac insects can be grown on several species, including A.
nilotica and A.
catechu. Some species have been used for forestation of wastelands, including A. nilotica, which can withstand some water inundation and a few such areas have become bird sanctuaries.

To date, approximately 330 compounds have been isolated from various Acacia species. Flavonoids, a type of water-soluble plant pigments, are the major class of compounds isolated from Acacias. Approximately 180 different flavonoids have been identified, 111 of which are flavans. Terpenoids are second largest class of compounds isolated from species of the Acacia genus, with 48 compounds having been identified.
Other classes of compounds isolated from Acacia include, alkaloids (28), amino acids/peptides (20), tannins (16), carbohydrates (15), oxygen heterocycles (15) and aliphatic compounds (10). (Buckingham, in The Combined Chemical Dictionary, Chapman & Hall CRC, version 5:2, Dec. 2001).
Phenolic compounds, particularly flavans are found in moderate to high concentrations in all Acacia species (Abdulrazak et al. (2000) J. Anim. Sci.
13:935-940).

Historically, most of the plants and extracts of the Acacia genus have been utilized as astringents to treat gastrointestinal disorders, diarrhea, indigestion and to stop bleeding (Vautrin (1996) Universite Bourgogne (France) European abstract 58-OIC:177;
Saleem et al. (1998) Hamdard Midicus. 41:63-67). The bark and pods of A. arabica Willd.
contain 5 large quantities of tannins and have been utilized as astringents and expectorants (Nadkarni (1996) India Materia Medica, Bombay Popular Prakashan, pp. 9-17).
Diarylpropanol derivatives, isolated from stem bark of A. tortilis from Somalia, have been reported to have smooth muscle relaxing effects (Hagos et al. (1987) Planta Med. 53:27-31, 1987). It has also been reported that terpenoid saponins isolated from A.
victoriae have 10 an inhibitory effect on dimethylbenz(a)anthracene-induced murine skin carcinogenesis (Hanausek et al. (2000) Proc. Am. Assoc. Can. Res. Annu. Mtg. 41:663) and induce apoptosis (Haridas et al. (2000) Proc. Am. Assoc. for Can. Res. Annu. Mtg.
41:600).
Plant extracts from A. nilotica have been reported to have spasmogenic, vasoconstrictor and anti-hypertensive activity (Amos et al. (1999) Phytotherapy Research 13:683-685;

15 Gilani et al. (1999) Phytotherapy Research 13:665-669), and antiplatelet aggregatory activity (Shah et al. (1997) Gen. Pharmacol. 29:251-255). Anti-inflammatory activity has been reported for A. nilotica. It was speculated that flavonoids, polysaccharides and organic acids were potential active components (Dafallah and Al-Mustafa (1996) Am. J.
Chin. Med. 24:263-269). To date, the only reported 5-lipoxygenase inhibitor isolated from Acacia is a monoterpenoidal carboxamide (Seikine et al. (1997) Chem. Pharm.
Bull.
(Tokyo) 45:148-11).
Acacia gums have been formulated with other plant ingredients and used for ulcer prevention without identification of any of the active components (Fuisz, U.S.
Pat. No.
5,651,987). Acacia gums have also been formulated with other plant ingredients and used to improve drug dissolution (Blank, U.S. Pat. No. 4,946,684), by lowering the viscosity of nutritional compositions (Chancellor, U.S. Pat. No. 5,545,411).

The extract from the bark of Acacia was patented in Japan for external use as a whitening agent (Abe, JP10025238), as a glucosyl transferase inhibitor for dental applications (Abe, JP07242555), as a protein synthesis inhibitor (Fukai, JP
07165598), as an active oxygen-scavenging agent for external skin preparations (Honda, JP
07017847, Bindra U.S. Pat. No. 6,1266,950), and as a hyaluronidase inhibitor for oral consumption to prevent inflammation, pollinosis and cough (Ogura, JP 07010768).
Review of the literature has revealed no human clinical applications using mixtures of Free-B-ring flavonoids and flavans for relief of pain or measuring biochemical clinical outcomes for osteoarthritis treatment. This report appears to be the first randomized, double blind, placebo controlled study of the safety and efficacy of these compounds in humans.

SUMMARY OF THE INVENTION
The present invention includes a novel composition of matter comprised of a mixture of Free-B-ring flavonoids and flavans. This novel composition of matter is referred to herein as Univestin". The ratio of Free-B-ring flavonoids to flavans in the composition of matter can be adjusted based on the indications and the specific requirements with respect to prevention and treatment of a specific disease or condition.
Generally, the ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.

The present invention father includes methods that are effective in simultaneously inhibiting both the COX-2 and 5-LO enzymes. The method for the simultaneous dual inhibition of the COX-2 and 5-LO pathways is comprised of administering a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants to a host in need thereof. The efficacy of this method was demonstrated with purified enzymes, in different cell lines, multiple animal models and eventually in a human clinical study. The ratio of Free-B-ring flavonoids to flavans in the composition can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment, the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.

The present invention further includes methods for the prevention and treatment of COX-2 and 5-LO mediated diseases and conditions, including but not limited to menstrual cramps, arteriosclerosis, heart attack, obesity, diabetes, syndrome X, Alzheimer's disease, respiratory allergic reaction, chronic venous insufficiency, hemorrhoids, Systemic Lupus Erythromatosis, psoriasis, chronic tension headache, migraine headaches, inflammatory bowl disease; topical infections caused by virus, bacteria and fungus, sunburn, thermal burns, contact dermatitis, melanoma and carcinoma.. The method for preventing and treating COX-2 and 5-LO mediated diseases and conditions is comprised of administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants together with a pharmaceutically acceptable carrier. The ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment, the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
In another embodiment, the present invention includes a method for treating general joint pain and stiffness, improving mobility and physical function and preventing and treating pathological conditions of osteoarthritis and rheumatoid arthritis. The method for preventing and treating joint pain and stiffness, improving mobility and physical function and preventing and treating pathological conditions of osteoarthritis, and rheumatoid arthritis comprises administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-13-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants together with a pharmaceutically acceptable carrier. The ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 to 1:99 Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment, the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
The present invention includes methods for weight loss and blood sugar control due to increased physical activity resulting from improving mobility, flexibility and physical function said method comprising administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants and a pharmaceutically acceptable carrier. The ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
The present invention also includes a method for modulating the production of mRNA implicated in pain pathways said method comprising administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants and a pharmaceutically acceptable carrier. While not limited by theory, Applicant believes that the ability to modulate the production of mRNA is accomplished via a decrease, by the active ingredients in the Free-B-ring/flavan composition, in the production of mRNA
by the cox-2 gene, but not the cox-1 gene. The ratio of Free-B-ring flavonoids to flavans in the composition can be in the range of 99:1 to 1:99 Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
The Free-B-ring flavonoids, also referred to herein as Free-B-ring flavones and flavonols, that can be used in accordance with the following invention.
include compounds illustrated by the following general structure:

A CI

wherein R,, R2, R3, R4, and Rs are independently selected from the group consisting of -H, -OH, -SH, OR, -SR, -NH2, -NHR, -NR2, -NR3+X-, a carbon, oxygen, nitrogen or sulfur, glycoside of a single or a combination of multiple sugars including, but not limited to aldopentoses, methyl-aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof;
wherein R is an alkyl group having between 1-10 carbon atoms; and X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, carbonate, etc.

The flavans that can be used in accordance with the following invention include compounds illustrated by the following general structure:

wherein 5 R1, R2, R3, R4 and R5 are independently selected from the group consisting of H, -OH, -SH, -OCH3, -SCH3, -OR, -SR, -NH2, -NRH, -NR2, -NR3+X-, esters of the mentioned substitution groups, including, but not limited to, gallate, acetate, cinnamoyl and hydroxyl-cinnamoyl esters, trihydroxybenzoyl esters and caffeoyl esters and their chemical derivatives thereof; carbon, oxygen, nitrogen or sulfur glycoside of a single or a 10 combination of multiple sugars including, but not limited to, aldopentoses, methyl aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof;
dimer, trimer and other polymerized flavans;

wherein R is an alkyl group having between 1-10 carbon atoms; and 15 X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, carbonate, etc.

The Free-B-ring flavonoids of this invention may be obtained by synthetic methods or extracted from the families of plants including, but not limited to Annonaceae, 20 Asteraceae, Bignoniaceae, Combretaceae, Compositae, Euphorbiaceae, Labiatae, Lauranceae, Leguminosae, Moraceae, Pinaceae, Pteridaceae, Sinopteridaceae, Ulmaceae and Zingiberaceae. The Free-B-ring flavonoids can be extracted, concentrated, and purified from the genera of high plants, including but not limited to Desmos, Achyrocline, Oroxylum, Buchenavia, Anaphalis, Cotula, Gnaphaliurn, Helichrysum, Centaurea, Eupatorium, Baccharis, Sapiurn, Scutellaria, Molsa, Colebrookea, Stachys, Origanum, Ziziphora, Lindera, Actinodaphne, Acacia, Derris, Glycyrrhiza, Millettia, Pongamia, Tephrosia, Artocarpus, Ficus, Pityrogranuna, Notholaena, Pinus, Ulmus and Alpinia.
As noted above the flavans of this invention may be obtained from a plant or plants selected from the genus of Acacia. In a preferred embodiment, the plant is selected from the group consisting of Acacia catechu, A. concinna, A. farnesiana, A.
Senegal, A.
speciosa, A. arabica, A. caesia, A. pennata, A. sinuata. A. mearnsii, A.
picnantha, A.
dealbata, A. auriculiformis, A. holoserecia and A. mangium.
The present invention includes an evaluation of different compositions of Free-B-ring flavonoids and flavans using enzymatic and in vivo models to optimize the formulation and obtain the best potency. The efficacy and safety of this formulation is also demonstrated in human clinical studies. The present invention provides a commercially viable process for the isolation, purification and combination of Acacia flavans with Free-B-ring flavonoids to yield composition of matter having desirable physiological activity.
The compositions of this invention can be administered by any method known to one of ordinary skill in the art. The modes of administration include, but are not limited to, enteral (oral) administration, parenteral (intravenous, subcutaneous, and intramuscular) administration and topical application. The method. of treatment according to this invention comprises administering internally or topically to a patient in need thereof a therapeutically effective amount of a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed.

BRIEF DESCRIPTION OF THE FIGURES
Figure 1 depicts graphically the inhibition of COX-1 and COX-2 by HTP
fractions from Acacia catechu. The extracts were prepared and fractionated as described in Examples 1 and 3. The extracts were examined for their inhibition of the peroxidase activity of recombinant ovine COX-1 (R) or ovine COX-2 (+) as described in Example 2.
The data is presented as percent of untreated control.

Figure 2 depicts graphically the inhibition of COX-1 and COX-2 by HTP
fractions from Scutellaria baicalensis. The extracts were prepared and fractionated as described in Examples 1 and 3. The extracts were examined for their inhibition of the peroxidase activity of recombinant ovine COX-1 (.) or ovine COX-2 (4) as described in Example 2.
The data is presented as percent of untreated control.
Figure 3 depicts a HPLC chromatogram of a standardized extract isolated from the roots of Scutellaria baicalensis (lot # RM052302-01) having a Free-B-ring flavonoid content of 82.2%. Ten structures were elucidated using HPLC/PDA/MS as baicalin, wogonin-7-glucuronide, oroxylin A 7-glucuronide, baicalein, wogonin, chrysin-7-glucuronide, norwogonin-7-glucuronide, scutellarin, chrysin and oroxylin A.

Figure 4 depicts graphically a profile of the inhibition of COX-1 and COX-2 by the baicalein, which was isolated and purified from Scutellaria baicalensis. The compound was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (=) and ovine COX-2 (.). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/mL). The IC50 for COX-1 was calculated as 0.18 g/mL/unit of enzyme while the IC50 for COX-2 was calculated as 0.28 g/mL/unit.

Figure 5 depicts graphically a profile of the inhibition of COX-1 and COX-2 by the baicalin, which was isolated and purified from Scutellaria baicalensis. The compound was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (4) and ovine COX-2 (a). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/mL). The IC50 for COX-1 was determined to be 0.44 g/mL/unit of enzyme while that of COX-2 was determined to be 0.28 pg/mL/unit.

Figure 6 depicts graphically a profile of the inhibition of COX-1 and COX-2 by a standardized Free-B-ring flavonoid extract (83% baicalin based on HPLC) isolated from Scutellaria baicalensis. The extract was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (+) and ovine COX-2 (a). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/mL). The IC50 for COX-1 was calculated as 0.24 g/mL/unit of enzyme while the IC50 for was calculated as 0.48 p.g/mL/unit.

Figure 7 depicts graphically a profile of the inhibition of COX-1 and COX-2 by catechin, which was isolated and purified from Acacia catechu. The compound was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (+) and ovine COX-2 (a). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/mL). The IC50 for COX-1 was determined to be 0.11 g/mL/unit of enzyme while the IC50 for COX-2 was determined as 0.42 g/mL/unit.
Figure 8 depicts graphically a profile of the inhibition of COX-1 and COX-2 by a standardized flavan extract containing 50% total catechins isolated from Acacia catechu.
The extract was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (4) and ovine COX-2 (a). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/ML). The IC50 for COX-1 was calculated as 0.17 g/mL/unit of enzyme while the ICS0 for COX-2 was determined to be 0.41 p g mL/unit.
Figure 9 depicts the HPLC chromatogram of the flavans extracted from Acacia catechu with 80% MeOH in water.

Figure 10 depicts graphically a profile of the inhibition of 5-LO by the purified flavan catechin from Acacia catechu. The compound was examined for its inhibition of recombinant potato 5-lipoxygenase activity (4). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/mL).
The IC50 for 5-LO was 1.38 g/rL/unit of enzyme.
Figure I l depicts graphically a profile of the inhibition of COX-1 and COX-2 by the UnivestinTM composition produced through combination of the extracts of Free-B-ring flavonoids and flavans in a ratio of 85:15 as described in Example 14.
UnivestinTM was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (4) and ovine COX-2 (.). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration (pg/mL). The IC50 for COX-1 was 0.76 gg/mL/unit of enzyme while the IC50 for COX-2 was 0.80 gg/mUunit.

Figure 12 depicts graphically a profile of the inhibition of COX-1 and COX-2 by the UnivestinTM composition produced through combination of Free-B-ring flavonoids and flavans extracts in a ratio of 50:50 as described in Example 14. UnivestinTM
was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (=) and ovine COX-2 (m). The data is presented as percent inhibition vs. inhibitor concentration ( g/mL). The IC50 for COX-1 was 0.38 g/mL/unit of enzyme while the IC50 for was 0.84 g/mL/unit.
Figure 13 depicts graphically a profile of the inhibition of COX-1 and COX-2 by the Univestin composition produced through combination extracts of Free-B-ring flavonoids and flavans in a ratio of 20:80 as described in Example 14.
Univestin was examined for its inhibition of the peroxidase activity of recombinant ovine COX-1 (+) and ovine COX-2 (.). The data is presented as percent inhibition of assays without inhibitor vs. inhibitor concentration ( g/mL). The IC50 for COX-1 was 0.18 g/mL/unit of enzyme while the IC50 for COX-2 was 0.41 g/mL/unit.

Figure 14 depicts the effect of increasing concentrations of UnivestinTM on the amount of LPS-induced newly synthesized LTB4 (4) as determined by ELISA in THP-1 or HT-29 cells (ATCC). The activity of the combination extract is expressed as %
inhibition of induced LTB4 synthesis.

Figure 15 compares the LTB4 levels as determined by ELISA that remain in HT-29 cells after treatment with 3 g/mL UnivestinTM in non-induced cells to treatment with 3 gg/mL ibuprofen as described in Example 16.

Figure 16 compares the effect of various concentrations of UnivestinTm on cox-and cox-2 gene expression. The expression levels are standardized to 18S rRNA
expression levels (internal control) and then normalized to the no-treatment, no-LPS
condition. This Figure demonstrates a decrease in cox-2, but not cox-1 gene expression following LPS-stimulation and exposure to UnivestinT.

Figure 17 compares the effect of 3p.g/mL UnivestinTM on cox-1 and cox-2 gene expression with the equivalent concentration of other NSAIDs. The expression levels are standardized to 18S rRNA expression levels (internal control) and then normalized to the no-treatment, no-LPS condition.

Figure 18 illustrates graphically ear-swelling data as a measure of inhibition of inflammation. UnivestinTM produced through the combination of standardized extracts of Free-B-ring flavonoids and flavans in a ratio of 80:20 was compared to untreated mice and mice given indomethacin (50 mg/kg) via oral gavage. The data is presented as the difference in micron measurement of the untreated vs. the treated ear lobe for each mouse.

Figure 19 shows the effect of 100 mg/kg of UnivestinTM (80:20) ratio of standardized extracts of Free-B-ring flavonoids to flavans) on the AA injected ankles of mice (UnivestinTM + arachidonic acid) compared to non-treated mice (no treatment +

arachidonic acid), mice without AA injections (negative control) or mice that were injected with the liquid carrier (vehicle control).
Figure 20 illustrates graphically the 95% confidence interval for the pain index WOMAC score at baseline, 30, 60 and 90 days of treatment with UnivestinTm at a dosage 5 of 250 mg/day.
Figure 21 illustrates graphically the 95% confidence interval for the pain index WOMAC score at baseline, 30, 60 and 90 days of treatment with UnivestinTm at a dosage of 500 mg/day.

Figure 22 illustrates graphically the 95% confidence interval for the pain index 10 WOMAC score at baseline, 30, 60 and 90 days of treatment with celecoxib at a dosage of 200 mg/day.

Figure 23 illustrates graphically the 95% confidence interval for the pain index WOMAC score at baseline, 30, 60 and 90 days of treatment with the placebo.

Figure 24 illustrates graphically the 95% confidence interval for the stiffness index 15 WOMAC score at baseline, 30, 60 and 90 days of treatment with UnivestinTM
at a dosage of 250 mg/day.
Figure 25 illustrates graphically the 95% confidence interval for the stiffness index WOMAC score at baseline, 30, 60 and 90 days of treatment with UnivestinTM at a dosage of 500 mg/day.

20 Figure 26 illustrates graphically the 95% confidence interval for the stiffness index WOMAC score at baseline, 30, 60 and 90 days of treatment with celecoxib at a dosage of 200 mg/day.

Figure 27 illustrates graphically the 95% confidence interval for the stiffness index WOMAC score at baseline, 30, 60 and 90 days of treatment with the placebo.
25 Figure 28 illustrates graphically the 95% confidence interval for the functional impairment index WOMAC score at baseline, 30, 60 and 90 days of treatment with UnivestinTM at a dosage of 250 mg/day.

Figure 29 illustrates graphically the 95% confidence interval for the functional impairment index WOMAC score at baseline, 30, 60 and 90 days of treatment with UnivestinTM at a dosage of 500 mg/day.
Figure 30 illustrates graphically the 95% confidence interval for the functional impairment index WOMAC score at baseline, 30, 60 and 90 days of treatment with celecoxib at a dosage of 200 mg/day.

Figure 31 illustrates graphically the 95% confidence interval for the functional impairment index WOMAC score at baseline, 30, 60 and 90 days of treatment with the placebo.

Figure 32 shows the effect of UnivestinTM at doses of 250 and 500 mg/day on decreasing BMI compared to celecoxib at 200 mg/day and the placebo.

Figure 33 shows the effect of UnivestinTM at doses of 250 and 500 mg/day on decreasing weight compared to celecoxib at 200 mg/day and the placebo.

Figure 34 shows the effect of UnivestinTM at doses of 250 and 500 mg/day on lowering blood glucose compared to placebo.

DETAILED DESCRIPTION OF THE INVENTION

Various terms are used herein to refer to aspects of the present invention. To aid in the clarification of the description of the components of this invention, the following definitions are provided.
It is to be noted that the term "a" or "an" entity refers to one or more of that entity;
for example, a flavonoid refers to one or more flavonoids. As such, the terms "a" or "an", "one or more" and "at least one" are used interchangeably herein.
"Free-B-ring Flavonoids" as used herein are a specific class of flavonoids, which have no substitute groups on the aromatic B ring, as illustrated by the following general structure:

IA ~I
3 o B

wherein R1, R2, R3, R4, and R5 are independently selected from the group consisting of -H, -OH, -SH, OR, -SR, -NH2, -NHR, -NR2, -NR3+X-, a carbon, oxygen, nitrogen or sulfur, glycoside of a single or a combination of multiple sugars including, but not limited to aldopentoses, methyl-aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof;

wherein R is an alkyl group having between 1-10 carbon atoms; and X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, carbonate, etc.

"Flavans" are a specific class of flavonoids, which can be generally represented by the following general structure:

s R1 )P&R3 wherein R1, R2, R3, R4 and R5 are independently selected from the group consisting of H, -OH, -SH, -OCH3, -SCH3, -OR, -SR, -NH2, -NRH, -NR2, -NR3+X esters of substitution groups, including, but not limited to, gallate, acetate, cinnamoyl and hydroxyl-cinnamoyl esters, trihydroxybenzoyl esters and caffeoyl esters and their chemical derivatives thereof;
carbon, oxygen, nitrogen or sulfur glycoside of a single or a combination of multiple sugars including, but not limited to, aldopentoses, methyl aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof; dimer, trimer and other polymerized flavans;

wherein R is an alkyl group having between 1-10 carbon atoms; and X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, carbonate, etc.

"Gene expression" refers to the transcription of a gene to mRNA.
"Protein expression" refers to the translation of mRNA to a protein.
"RT-qPCR" is a method for reverse transcribing (RT) an mRNA molecule into a cDNA molecule and then quantitatively evaluating the level of gene expression using a polymerase chain reaction (PCR) coupled with a fluorescent reporter.

"Therapeutic" as used herein, includes treatment and/or prophylaxis. When used, therapeutic refers to humans as well as other animals.

"Pharmaceutically or therapeutically effective dose or amount" refers to a dosage level sufficient to induce a desired biological result. That result may be the alleviation of the signs, symptoms or causes of a disease or any other alteration of a biological system that is desired.
"Placebo" refers to the substitution of the pharmaceutically or therapeutically effective dose or amount sufficient to induce a desired biological that may alleviate the signs, symptoms or causes of a disease with a non-active substance.

A "host" or "patient" is a living subject, human or animal, into which the compositions described herein are administered.
Note that throughout this application various citations are provided. Each citation is specifically incorporated herein in its entirety by reference.
The present invention includes a novel composition of matter comprised of a mixture of Free-B-ring flavonoids and flavans. This novel composition of matter is referred to herein as UnivestinTM. The ratio of Free-B-ring flavonoids to flavans in the composition of matter can be adjusted based on the indications and the specific requirements with respect to prevention and treatment of a specific disease or condition.
Generally, the ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
In one embodiment of the present invention, the standardized Free-B-ring flavonoid extract is comprised of the active compounds with a purity of between 1-99%
(by weight) of total Free-B-ring flavonoids as defined in examples 5, 7 and 13; Tables 5, 7, 8 and 9 and Figure 3. Baicalin is the major active component in the extract, which accounts for approximately 50-90% (by weight) of the total Free-B-ring flavonoids. In a preferred embodiment, the standardized extract contains >70% total Free-B-ring flavonoids in which >75% of the Free-B-ring flavonoids is baicalin.
In one embodiment, the standardized flavan extract is comprised of the active compounds with a purity of between 1-99% (by weight) total flavans as defined in Example 8, 9 and 12; Tables 4, 6 and 9 and Figure 9. Catechin is the major active component in the extract and accounts for 50-90% (by weight) of the total flavans. In a preferred embodiment, the standardized flavan extract contains >50% total flavans in which >70% of flavans is catechin.
In one embodiment UnivestinTM is be produced by mixing the above two extracts or synthetic compounds in a ratio from 99:1 to 1:99. The preferred ratio of Free-B-ring flavonoids to flavans is 85:15 Free-B-ring flavonoids:flavans as defined in Example 14.
The concentration of Free-B-ring flavonoids in UnivestinTM can be from about 1%
to 99% and the concentration of flavans in UnivestinTM can be from 99% to 1%.
In a preferred embodiment of the invention, the concentration of total Free-B-ring flavonoids in UnivestinTM is approximately 75% with a baicalin content of approximately 60%
of total weight of the UnivestinTM; and the concentration of total flavans in UnivestinTM is approximately 10% with a catechin content of approximately 9%. In this embodiment, the total active components (Free-B-ring flavonoids plus flavans) in UnivestinTM
are >80% of the total weight.
The present invention also includes methods that are effective in simultaneously inhibiting both the COX-2 and 5-LO enzymes. The method for the simultaneous dual inhibition of the COX-2 and 5-LO pathways is comprised of administering a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants to a host in need thereof. The ratio of Free-B-ring flavonoids to flavans in the composition can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred 5 embodiment, the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
The present further includes methods for the prevention and treatment of COX-2 and 5-LO mediated diseases and conditions. The method for preventing and treating 10 COX-2 and 5-LO mediated diseases and conditions is comprised of administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants together with a pharmaceutically acceptable carrier. The ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 Free-B-ring flavonoids:flavans to 1:99 of Free-B-ring 15 flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment, the Free-B-ring flavonoids are isolated 20 from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
In yet a further embodiment, the present includes a method for treating general joint pain and stiffness, improving mobility and physical function and preventing and treating pathological conditions of osteoarthritis and rheumatoid arthritis.
The method for 25 preventing and treating joint pain and stiffness, improving mobility and physical function and preventing and treating pathological conditions of osteoarthritis, and rheumatoid arthritis is comprised of by administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants together with a pharmaceutically 30 acceptable carrier. The'ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 to 1:99 Free-B-ring flavonoids:flavans. In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment, the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.

The present invention also includes a method for modulating the production of mRNA implicated in pain pathways said method comprising administering to a host in need thereof an effective amount of a composition comprising a mixture of Free-B-ring flavonoids and flavans synthesized and/or isolated from a single plant or multiple plants and optionally a pharmaceutically acceptable carrier. The ratio of Free-B-ring flavonoids to flavans can be in the range of 99:1 to 1:99 Free-B-ring flavonoids:flavans.
In specific embodiments of the present invention, the ratio of Free-B-ring flavonoids to flavans is selected from the group consisting of approximately 90:10, 80:20, 70:30, 60:40, 50:50, 40:60, 30:70, 20:80 and 10:90. In a preferred embodiment of the invention, the ratio of Free-B-ring flavonoids:flavans in the composition of matter is approximately 85:15. In a preferred embodiment the Free-B-ring flavonoids are isolated from a plant or plants in the Scutellaria genus of plants and flavans are isolated from a plant or plants in the Acacia genus of plants.
The Free-B-ring flavonoids that can be used in accordance with the method of this invention include compounds illustrated by the general structure set forth above. The Free-B-ring flavonoids of this invention may be obtained by synthetic methods or may be isolated from the family of plants including, but not limited to Annonaceae, Asteraceae, Bignoniaceae, Combretaceae, Coinpositae, Euphorbiaceae, Labiatae, Lauranceae, Leguminosae, Moraceae, Pinaceae, Pteridaceae, Sinopteridaceae, Ulmaceae, and Zingiberaceae. The Free-B-ring flavonoids can also be extracted, concentrated, and purified from the genera of high plants, including but not limited to Desmos, Achyrocline, Oroxylum, Buchenavia, Anaphalis, Cotula, Gnaphalium, Helichrysum, Centaurea, Eupatorium, Baccharis, Sapium, Scutellaria, Molsa, Colebrookea, Stachys, Origanum, Ziziphora, Lindera, Actinodaphne, Acacia, Derris, Glycyrrhiza, Millettia, Pongamia, Tephrosia, Artocarpus, Ficus, Pityrogramma, Notholaena, Pinus, Ulrnus, and Alpinia.

The Free-B-ring flavonoids can be found in different parts of plants, including but not limited to stems, stem barks, twigs, tubers, roots, root barks, young shoots, seeds, rhizomes, flowers and other reproductive organs, leaves and other aerial parts. Methods for the isolation and purification of Free-B-ring flavonoids are described in U.S.
Application Serial No. 10/091,362, filed March 1, 2002, entitled "Identification of Free-B-ring Flavonoids as Potent COX-2 Inhibitors," which is incorporated herein by reference in its entirety.

The flavans that can be used in accordance with the method of this invention include compounds illustrated by the general structure set forth above. The flavans of this invention may be obtained by synthetic methods or may be isolated from a plant or plants selected from the Acacia genus of plants. In a preferred embodiment, the plant is selected from the group consisting of Acacia catechu, A. concinna, A. farnesiana, A.
Senegal, A.

speciosa, A. arabica, A. caesia, A. pennata, A. sinuata. A. mearnsii, A.
picnantha, A.
dealbata, A. auriculiformis, A. holoserecia and A. mangium.

The flavans can be found in different parts of plants, including but not limited to stems, stem barks, trunks, trunk barks, twigs, tubers, roots, root barks, young shoots, seeds, rhizomes, flowers and other reproductive organs, leaves and other aerial parts. Methods for the isolation and purification of flavans are described in U.S.
Application Serial No.
10/104,477, filed March 22, 2002, entitled "Isolation of a Dual COX-2 and 5-Lipoxygenase Inhibitor from Acacia," which is incorporated herein by reference in its entirety.

The present invention implements a strategy that combines a series of in vivo studies as well as in vitro biochemical, cellular, and gene expression screens to identify active plant extracts and components that specifically inhibit COX-2 and 5-LO
enzymatic activity, and impact cox-2, but not cox-1 mRNA production. The methods used herein to identify active plant extracts and components that specifically inhibit COX-2 and 5-LO
pathways are described in Examples 1 to 13 (Figures 1-10). These methods are described in greater detail in U.S. Application Serial No. 10/091,362, filed March 1, 2002, entitled "Identification of Free-B-ring Flavonoids as Potent COX-2 Inhibitors" and U.S.
Application Serial No. 10/104,477, filed March 22, 2002, entitled "Isolation of a Dual COX-2 and 5-Lipoxygenase Inhibitor from Acacia," each of which is specifically incorporated herein by reference in its entirety.
These studies resulted in the discovery of a novel composition of matter referred to herein as UnivestinTM, which is comprised of a proprietary blending of two standardized extracts, which contain Free-13-ring flavonoids and flavans, respectively. A
general example for preparing such a composition is provided in Example 14 using two standardized extracts isolated from Acacia and Scutellaria, respectively, together with one or more excipients. The Acacia extract used in Example 14 contained >60% total flavans, as catechin and epicatechin, and the Scutellaria extract contained >70% Free-B-ring flavonoids, which was primarily baicalin. The Scutellaria extract contained other minor amounts of Free-B-ring flavonoids as set forth in Table 11. One or more excipients are optionally added to the composition of matter. The amount of excipient added can be adjusted based on the actual active content of each ingredient desired. A
blending table for each individual batch of product must be generated based on the product specification and QC results for individual batch of ingredients. Additional amounts of active ingredients in the range of 2-5% are recommended to meet the product specification. Example illustrates a blending table that was generated for one batch of Univestin (Lot#G1702-COX-2). Different blending ratios of the formulated Univestin' product were tested for their ability to inhibit COX-2 and 5-LO enzyme activities, and to reduce cox mRNA
production as described in Examples 15-17.

The COX-2 inhibition assay relied on the activity of the enzyme peroxidase in the presence of heme and arachidonic acid. In order to screen for compounds that inhibited COX-1 and COX-2 activity, a high throughput, in vitro assay was developed that utilized inhibition of the peroxidase activity of both enzymes as illustrated in Examples 2 and 6.
After isolating plant fractions that inhibited COX-2 activity in the screening process, the two individual standardized extracts, one composed primarily of Free-B-ring flavonoids (isolated from Scutellaria) and the other of flavans (isolated from Acacia), were compared, as well as, purified components from each extract and different ratios of the combined extracts by titrating against a fixed amount of the COX-1 and COX-2 enzymes.
This study revealed that the purified Free-B-ring flavonoids, baicalin and baicalein isolated from Scutellaria baicalensis and the purified flavan, catechin isolated from Acacia catechu, inhibited COX-2 and 5-LO activity. Additionally, each of the individual standardized extracts, which contained concentrations of Free-B-ring flavonoids in the range of 10-90%
(based on HPLC) and flavans in the range of 10-90% (based on HPLC), also inhibited COX-2 and 5-LO activity. Finally, the study revealed that compositions containing mixtures of each of the individual standardized extracts having ratios of Free-B-ring flavonoids to flavans of approximately 80:20, 50:50, and 20:80, were also all highly effective at inhibiting COX-2 enzymatic activity in vitro. The results are set forth in Figures 11-13).
Example 16 describes cell assays performed that targeted inhibition of compounds in the breakdown of arachidonic acid in the 5-LO pathway, namely LTB4. The results are set forth in Figures 14 and 15.
Example 17 describes an experiment performed to determine differential inhibition of the cox-2 gene by UnivestinTm. Gene expression data was obtained for the inhibition of cox-1 and cox-2 mRNA production in a semi-quantitative RT-qPCR assay. The results are set forth in Figures 16 and 17. With reference to Figure 16 it can be seen that UnivestinTM
inhibited cox-2 mRNA production without effecting cox-1 gene expression. In addition, when compared with other COX-2 inhibitor drugs, UnivestinTm was able to decrease LPS-stimulated increases in cox-1 and cox-2 gene expression. Importantly, celecoxib and ibuprofen both increased cox-2 gene expression (Figure 17).
In vivo efficacy was demonstrated by the application of skin irritating substances, such as AA, to the ears of mice and measuring the reduction of swelling in mice treated with UnivestinTM as described in Example 18. The results are set forth in Figure 18.
Additionally, efficacy at the site of inflammation and pain, was determined by the injection of an irritant into the ankle joints of mice and measuring the reduction of swelling in mice treated with UnivestinTM, as described in Example 19. The results are set forth in Figure 19.
Individual standardized extracts containing concentrations of Free-B-ring flavonoids in the range of 10-99% (based on HPLC) and flavans in the range of 10-99%
(based on HPLC) as well as the product UnivestinTM were tested for toxicity in mice with chronic and acute administration (data not shown). In the chronic administration protocol, mice were fed the test articles by oral gavage with daily doses of 90 mg/kg (equivalent to the human daily dose of 500 mg), 450 mg/kg (five times the daily-dose equivalent) and 900 mg/kg (ten times the daily-dose equivalent). Mice showed no adverse effects in terms of weight gain, physical appearance or behavior. Gross necropsy results showed no organ 5 abnormalities and histology of the stomach, kidney, and liver showed no differences compared to untreated control mice. Full blood work measuring electrolytes, blood proteins, blood enzymes, and liver enzymes showed no abnormalities compared to the untreated control mice. In the acute protocol, individual standardized extracts containing concentrations of Free-B-ring flavonoids in the range of 10-99% (based on HPLC) and 10 flavans in the range of 10-99% (based on HPLC) as well as the product UnivestinTM given at 2 grams/kg (20 times the daily-dose equivalent) showed no abnormalities in weight gain, appearance, behavior, gross necropsy appearance of organs, histology of stomach, kidney, and liver or blood work.

Example 20 describes a clinical study performed to evaluate the efficacy of 15 UnivestinTM on the relief of pain caused by rheumatoid arthritis or osteoarthritis of the knee and/or hip. The study was a single-center, randomized, double-blind, placebo-controlled study. Sixty subjects (n=60) with rheumatoid arthritis or osteoarthritis of the knee and/or hip were randomly placed into four groups and treated for 90 days with a placebo, UnivestinTM (250 mg/day or 500 mg/day) or CelebrexTM (also known as 20 celecoxib) (200 mg/day). The UnivestinTm, as illustrated in Example 14, Table 11, consisted of a proprietary blend of standardized extract of Scutellaria baicalensis Georgi with a baicalin content of 82.2% (w/w) and total Free-B-ring Flavonoids >90%
(w/w) and a standardized extract of Acacia catechu with a total flavan content of 77.2%
(w/w) in a ratio of 85:15. CelebrexTM is a trade name for a prescription drug that is a COX-2 selective 25 inhibitor. Table 12 sets forth the WOMAC index scores for pain, stiffness and function before treatment (baseline scores) and at 30, 60 and 90 days. Table 13 sets forth the absolute changes in WOMAC index scores for pain, stiffness and function after treatment for 30, 60 and 90 days. Figures 20-31 illustrate the results of this study graphically plotting the 95% confidence intervals for all data.
30 As shown in the Figures 20 to 31, the WOMAC composite scores and individual subscores, related to pain, stiffness and physical function exhibited significant improvements during administration of UnivestinTM compared to the placebo group.
Further, UnivestinTM exhibited a similar effectiveness on pain relieve, better effectiveness at decreasing stiffness, and marked improvement of physical function compared to the prescription drug CelebrexTM. The greatest significance can be seen in comparing each dose of UnivestinTM to the placebo and celecoxib in relieving pain, stiffness and functional impairment associated with osteoarthritis or rheumatoid arthritis.
Multiple post-hoc comparisons for each treatment group pairs within the Analysis of Variance models showed that UnivestinTM at 500 mg/day was significantly more effective than celecoxib at 200 mg/day for the reduction of pain caused osteoarthritis during the 30 days (p=0.020) of treatment. In addition, the administration of a dose of 500 mg/day of UnivestinTM was also significantly more effective than the placebo for the reduction of pain within 30 days (p=0.044), 60 days (p=0.032) and 90 days (p=0.001) of treatment. Celecoxib at 200 mg/day showed significance for the reduction of pain vs. the placebo at 60 days (p=0.009) of treatment. At 90 days, the 500 mg/day UnivestinTM dose showed significantly higher effectiveness compared to the 250 mg/day dose within 90 days (p=0.038) of treatment.

UnivestinTM at 250 mg/day was significantly more effective than the placebo for the reduction of stiffness caused by osteoarthritis, within 30 days (p=0.00), 60 days (p=0.027) and 90 days (p=0.015) of treatment. In addition, UnivestinTM at a dose of 500 mg/day was significantly more effective than placebo for reduction of stiffness caused by osteoarthritis, within 30 days (p=0.001) and 90 days (p=0.005) of treatment. Celecoxib at 200 mg/day showed significantly more effectiveness than the placebo for the reduction of stiffness caused by osteoarthritis only at 30 days (p=0.023) of treatment.

For reduction of functional impairment caused by osteoarthritis, UnivestinTM
was significantly more effective than celecoxib at 200 mg/day within 30 days (p=0.010) of treatment. In addition, the 250 mg/day dose of UnivestinTM was also significantly more effective than placebo for the reduction of functional impairment caused by osteoarthritis within 30 days (p=0.010), 60 days (p=0.043) and 90 days (p=0.039) of treatment. The 500 mg/day dose of UnivestinTM was more effective than celecoxib at 200 mg/day within 30 days (p=0.015), 60 days (p=0.043) and 90 days (0.039) of treatment. Finally, the 500 mg/day dose of UnivestinTM was also significantly more effective than placebo for the reduction of functional impairment caused by osteoarthritis within 30 days (p=0.015), 60 days (p=0.016) and 90 days (p=0.003) of treatment.

These results suggest that UnivestinTM, particularly at a dosage of 500 mg/day, is much more effective than the placebo and celecoxib at relieving pain, stiffness and improving functional impairment caused by osteoarthritis. Additionally, UnivestinTM

administered at a dosage of 250 mg/day is also very effective at relieving stiffness and improving functional impairment caused by osteoarthritis compared to the placebo and celecoxib. Celecoxib also showed only marginal improvement overall in relieving pain, stiffness and functional impairment caused by osteoarthritis.

In addition to the effects of UnivestinTM on pain, stiffness and functional impairment caused by osteoarthritis, Example 21 shows a measurable effect by UnivestinTm on body mass index (BMI) and weight loss. While not limited by theory, this effect may be due to an increase in mobility as a result of the administration of an anti-inflammatory or may also be due to a specific mechanism that increases metabolism or reduces the utilization of fats and carbohydrates in the body. Table 14 shows the effect of UnivestinTM administered at a dose of 250 and 500 mg/day as well as celecoxib and placebo on weight and BMI after 30 and 90 days of treatment. The results are illustrated graphically in Figures 32 and 33. With reference to Figures 32 and 33, it can be seen that Univestin administered at a dosage of both 250 and 500 ing/day resulted in a significant drop in weight and BMI after thirty days, with weight loss almost doubling after 90 days.
Celecoxib had a smaller effect on weight and BMI as compared to UnivestinTM.

Multiple post-hoc comparisons for each treatment group pairs with the Analysis of Variance models were also performed for weight loss and BMI as described in Example 21. These analyses showed that UnivestinTm at 250 mg/day and 500 mg/day doses caused statistically significant weight loss (p=0.011 vs. p=0.118) against the placebo after 30 days of treatment. Celecoxib did not cause significant weight loss against placebo at 30 days.
The weight loss continued throughout 90 days of treatment with Univestin at 250 and 500 mg/day with statistical significance versus placebo (p=0.001 and 0.01 receptively).
Celecoxib still did not show significance relative to the placebo. The decrease of BMI
followed similar trends for the 250 mg/day dose of Univestin which was significant relative to the placebo after 30 days (p=0.008) as well as 90 days (p=0.001).
The 500 mg/day dose of Univestin showed decreasing of BMI without statistical significance at 30 days of treatment. However, the decrease of BMI reached statistical significance 90 days of treatment (p=0.011). Again, after 90 days of treatment, the celecoxib treatment group showed no statistically significant changes in BMI versus placebo.

Example 22 suggests that administration of UnivestinTM may affect blood glucose levels as well as its effect on weight loss and BMI. Measurable differences in blood glucose levels are detected with 30 days of initiating treatment with UnivestinTm. At 90 days, both the 250 and 500 mg/day UnivestinTM treated groups showed significant drops in blood glucose levels. The effect of celecoxib on blood glucose was less dramatic. The results are set forth in Table 15 and illustrated graphically in Figure 34.
Once again multiple post-hoc comparisons for each treatment group pairs with the Analysis of Variance models were also performed for blood glucose as described in Example 22. Only the 500 mg/day dose of Univestin showed statistically relevant significance versus the placebo group (after30 days, p=0.028; after 90 days, p=0.022). T he 250 mg/day dose of Univestin, however, showed clinically significant changes in blood glucose levels versus the placebo.
The applicant believes that U.S. Application Serial No. 10/104,477, filed March 22, 2002, entitled "Isolation of a Dual COX-2 and 5-Lipoxygenase Inhibitor from Acacia,"
is the first report of a composition of matter isolated from the Acacia genus of plants that demonstrates dual specificity for COX-2 and 5-LO and that U.S. Application Serial No.
10/091,362, filed March 1, 2002, entitled "Identification of Free-B-ring Flavonoids as Potent COX-2 Inhibitors," is the first report of a correlation between Free-B-ring flavonoid structure and COX-2 inhibitory activity. These discoveries led to a novel blending of two classes of specific compounds --Free-B-ring flavonoids and flavans--to produce a composition of matter, referred to herein as UnivestinTM, which can be used for alleviating joint pain and stiffness, improving mobility and physical function and preventing and treating the pathological conditions of osteoarthritis, and rheumatoid arthritis.
While not limited by theory, the identified mechanism of action of this formulation is believed to be the direct inhibition of both the peroxidase activity of the COX-2 enzyme and the 5-LO enzyme activity, together with a decrease in the mRNA production of each of these enzymes. UnivestinTM can also be utilized to prevent and treat COX-2 and 5-LO

mediated diseases and conditions, including, but are not limited to osteoarthritis, rheumatoid arthritis, menstrual cramps, arteriosclerosis, heart attack, obesity, diabetes, syndrome X, Alzheimer's disease, respiratory allergic reaction, chronic venous insufficiency, hemorrhoids, Systemic Lupus Erythromatosis, psoriasis, chronic tension headache, migraine headaches, inflammatory bowl disease; topical infections caused by virus, bacteria, fungus, sunburn, thermal burns, contact dermatitis, melanoma and carcinoma. Finally, UnivestinTM has been found in human clinical study that it can cause weight loss and reduce blood glucose level due to improvement of flexibility, mobility and increase physical activity.

The present invention is also directed toward therapeutic compositions comprising the therapeutic agents of the present invention. The therapeutic agents of the instant invention can be administered by any suitable means, including, for example, parenteral, topical, oral or local administration, such as intradermally, by injection, or by aerosol. The particular mode of administration will depend on the condition to be treated.
It is contemplated that administration of the agents of the present invention may be via any bodily fluid, or any target or any tissue accessible through a body fluid. In the preferred embodiment of the invention, the agent is administered by injection. Such injection can be locally administered to any affected area. A therapeutic composition can be administered in a variety of unit dosage forms depending upon the method of administration.
For example, unit dosage forms suitable for oral administration of an animal include powder, tablets, pills and capsules. Preferred delivery methods for a therapeutic composition of the present invention include intravenous administration and local administration by, for example, injection or topical administration. A therapeutic reagent of the present invention can be administered to any animal, preferably to mammals, and more preferably to humans.
For particular modes of delivery, a therapeutic composition of the present invention can be formulated so as to include other components such as a pharmaceutically acceptable excipient, an adjuvant, and/or a carrier. For example, compositions of the present invention can be formulated in an excipient that the animal to be treated can tolerate.
Examples of such excipients, include but are not limited to cellulose, silicon dioxide, dextrates, sucrose, sodium starch glycolate, calcium phosphate, calcium sulfate, water, saline, Ringer's solution, dextrose solution, mannitol, Hank's solution, and other aqueous physiologically balanced salt solutions. Nonaqueous vehicles, such as fixed oils, sesame oil, ethyl oleate, or triglycerides may also be used. Other useful formulations include suspensions containing viscosity-enhancing agents, such as sodium 5 carboxymethylcellulose, sorbitol, or dextran. Excipients can also contain minor amounts of additives, such as substances that enhance isotonicity and chemical stability. Examples of buffers include phosphate buffer, bicarbonate buffer, Tris buffer, histidine, citrate, and glycine, or mixtures thereof, while examples of preservatives include thimerosal, m- or o-cresol, formalin and benzyl alcohol. Standard formulations can either be liquid injectables 10 or solids, which can be taken up in a suitable liquid as a suspension or solution for injection. Thus, in a non-liquid formulation, the excipient can comprise dextrose, human serum albumin, preservatives, etc., to which sterile water or saline can be added prior to administration.

In one embodiment of the present invention, the composition can also include an 15 adjuvant or a carrier. Adjuvants are typically substances that generally enhance the function of the formula in preventing and treating indications related to COX
& LO
pathways. Suitable adjuvants include, but are not limited to, Freund's adjuvant; other bacterial cell wall components; aluminum-based salts; calcium-based salts;
silica; boron, histidine, glucosamine sulfates, Chondroitin sulfate, copper gluconate, polynucleotides;

20 vitamin D, vitamin K, toxoids; shark and bovine cartilage; serum proteins;
viral coat proteins; other bacterial-derived preparations; gamma interferon; block copolymer adjuvants, such as Hunter's Titermax adjuvant (Vaxcel.TM., Inc. Norcross, Ga.); Ribi adjuvants (available from Ribi ImmunoChem Research, Inc., Hamilton, Mont.);
and saponins and their derivatives, such as Quil A (available from Superfos Biosector A/S, 25 Denmark). Carriers are typically compounds that increase the half-life of a therapeutic composition in the treated animal. Suitable carriers include, but are not limited to, polymeric controlled release formulations, biodegradable implants, liposomes, bacteria, viruses, oils, esters, and glycols.
One embodiment of the present invention is a controlled release formulation that is 30 capable of slowly releasing a composition of the present invention into an animal. As used herein, a controlled release formulation comprises a composition of the present invention in a controlled release vehicle. Suitable controlled release vehicles include, but are not limited to, biocompatible polymers, other polymeric matrices, capsules, microcapsules, microparticles, bolus preparations, osmotic pumps, diffusion devices, liposomes, lipospheres, and transdermal delivery systems. Other controlled release formulations of the present invention include liquids that, upon administration to an animal, form a solid or a gel in situ. Preferred controlled release formulations are biodegradable (i.e., bioerodible).

Once the therapeutic composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder;
or directly capsulated and/or tableted with other inert carriers for oral administration. Such formulations may be stored either in a ready to use form or requiring reconstitution immediately prior to administration. The manner of administering formulations containing the compositions for systemic delivery may be via oral, subcutaneous, intramuscular, intravenous, intranasal or vaginal or rectal suppository.
The amount of the composition that will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder of condition, which can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness or advancement of the disease or condition, and should be decided according to the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curved derived from in vitro or animal model test systems.
For example, an effective amount of the composition can readily be determined by administering graded doses of the composition and observing the desired effect.

The method of treatment according to this invention comprises administering internally or topically to a patient in need thereof a therapeutically effective amount of the composition comprised of a mixture of Free-B-ring flavonoids and flavans. The purity of the mixture includes, but is not limited to 0.01% to 100%, depending on the methodology used to obtain the compound(s). In a preferred embodiment, doses of the mixture of Free-B-ring flavonoids and flavans and pharmaceutical compositions containing the same are an efficacious, nontoxic quantity generally selected from the range of 0.01 to 200 mg/kg of body weight. Persons skilled in the art using routine clinical testing are able to determine optimum doses for the particular ailment being treated.

The following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention.

EXAMPLES
Example 1. Preparation of Organic and Aqueous Extracts from Acacia and Scutellaria Plants Plant material from Acacia catechu (L) Willd. barks, Scutellaria orthocalyx roots, Scutellaria baicalensis roots or Scutellaria lateriflora whole plant was ground to a particle size of no larger than 2 mm. Dried ground plant material (60 g) was then transferred to an Erlenmeyer flask and methanol:dichloromethane (1:1) (600 mL) was added. The mixture was shaken for one hour, filtered and the biomass was extracted again with methanol:
dichloromethane (1:1) (600 mL). The organic extracts were combined and evaporated under vacuum to provide the organic extract (see Table 1 below). After organic extraction, the biomass was air dried and extracted once with ultra pure water (600 mL).
The aqueous solution was filtered and freeze-dried to provide the aqueous extract (see Table 1 below).
Table 1. Yield of Organic and Aqueous Extracts of Acacia and Scutellaria Species Plant Source Amount Organic Extract Aqueous Extract Acacia catechu barks -60_g 27.2 g 10.8 Scutellaria orthocalyx roots 60 g 4.04 g 8.95 g Scutellaria baicalensis roots 60 g 9.18 7.18 g Scutellaria lateriflora 60 g 6.54 g 4.08 g (whole plant) Example 2. Inhibition of COX-2 and COX-1 Peroxidase Activity by Plant Extracts from Acacia catechu, Various Scutellaria Species and Other Plants The bioassay directed screening process for the identification of specific COX-inhibitors was designed to assay the peroxidase activity of the enzyme as described below.
Peroxidase Assay. The assay to detect inhibitors of COX-2 was modified for a high throughput platform (Raz). Briefly, recombinant ovine COX-2 (Cayman) in peroxidase buffer (100 mM TBS, 5 mM EDTA, 1 p.M Herne, 1 mg epinephrine, 0.094%
phenol) was incubated with extract (1:500 dilution) for 15 minutes. Quantablu (Pierce) substrate was added and allowed to develop for 45 minutes at 25 C.
Luminescence was then read using a Wallac Victor 2 plate reader. The results are presented in Table 2.
Table 2 sets forth the inhibition of enzyme by the organic and aqueous extracts obtained from five plant species, including the bark of Acacia catechu, roots of two Scutellaria species and extracts from three other plant species, which are comprised of structurally similar Free-B-ring flavonoids. Data is presented as the percent of peroxidase activity relative to the recombinant ovine COX-2 enzyme and substrate alone.
The percent inhibition by the organic extract ranged from 30% to 90%.

Table 2. Inhibition of COX-2 Peroxidase activity by various s ecies Plant Source Inhibition of COX-2 Inhibition of COX-2 b or anic extract by aqueous extract Acacia catechu (bark) 75% 30%
Scutellaria orthocalyx (root) 55% 77%
Scutellaria baicalensis (root) 75% 0%
Desmodium sambuense (whole plant) 55% 39%
Eucaluptus globulus (leaf) 30% 10%
Murica nana (leaf) 90% 0%

Comparison of the relative inhibition of the COX-1 and COX-2 isoforms requires the generation of IC50 values for each of these enzymes. The IC50 is defined as the concentration at which 50% inhibition of enzyme activity in relation to the control is achieved by a particular inhibitor. In these experiments, IC50 values were found to range from 6 to 50 g/mL and 7 to 80 g/mL for the COX-2 and COX-1 enzymes, respectively, as set forth in Table 3. Comparison of the IC50 values of COX-2 and COX-1 demonstrates the specificity of the organic extracts from various plants for each of these enzymes. The organic extract of Scutellaria lateriflora for example, shows preferential inhibition of COX-2 over COX-1 with IC50 values of 30 and 80 pg/rnL, respectively. While some extracts demonstrate preferential inhibition of COX-2, others do not.
Examination of the HTP fractions and purified compounds from these fractions is necessary to determine the true specificity of inhibition for these extracts and compounds.

Table 3. IC50 Values of Organic Extracts for Human and Ovine COX-2 and COX-1 Plant Source IC50 Human IC50 Ovine IC50 Ovine /mL ( mL mL
Acacia catechu (bark) 3 6.25 2.5 Scutellaria orthocalyx (root) Not done 10 10 Scutellaria baicalensis (root) 30 20 20 Scutellaria laterflora (whole plant) 20 30 80 Eucaluptus globulus (leaf) Not done 50 50 Murica nana (leaf) 5 6 7 Example 3. HTP Fractionation of Active Extracts Organic extract (400 mg) from active plant was loaded onto a prepacked lia~il flash column. (2 cm ID x 8.2 cm, lOg silica gel). The column was eluted using a Hitachi high throughput purification (HTP) system with a gradient mobile phase of (A) 50:50 EtOAc:hexane and (B) methanol from 100% A to 100% B in 30 minutes at a flow rate of 5 mL/min. The separation was monitored using a broadband wavelength UV detector and the fractions were collected in a 96-deep-well plate at 1.9 mL/well using a Gilson fraction collector. The sample plate was dried under low vacuum and centrifugation.
DMSO (1.5 mL) was used to dissolve the samples in each cell and a portion (100 L was taken for the COX inhibition assay.
Aqueous extract (750 mg) from active plant was dissolved in water (5 mL), filtered through a 1 pm syringe filter and transferred to a 4 mL High Pressure Liquid Chromatography (HPLC) vial. The solution was then injected by an autosampler onto a prepacked reverse phase column (C-18, 15 m particle size, 2.5 cm ID x 10 cm with precolumn insert). The column was eluted using a Hitachi high throughput purification (HTP) system with a gradient mobile phase of (A) water and (B) methanol from 100% A
to 100% B in 20 minutes, followed by 100% methanol for 5 minutes at a flow rate of 10 mL/min. The separation was monitored using a broadband wavelength UV detector and the fractions were collected in a 96-deep-well plate at 1.9 mL/well using a Gilson fraction collector. The sample plate was freeze-dried. Ultra pure water (1.5 mL) was used to dissolve samples in each cell and a portion (100 L) was taken for the COX
inhibition assay.

Example 4. Inhibition of COX Peroxidase Activity by HTP Fractions from Acacia and Scutellaria Species Individual bioactive organic extracts were further characterized by examining each of the HTP fractions for the ability to inhibit the peroxidase activity of both COX-1 and 5 COX-2 recombinant enzymes. The results are presented in Figures 1 and 2, which depict the inhibition of COX-2 and COX-1 activity by HTP fractions from organic extracts of the bark of Acacia catechu and the roots of Scutellaria baicalensis isolated as described in Examples 1 and 3 and assayed as described in Example 2. The profiles depicted in Figures 1 and 2 show multiple peaks of inhibition that indicate multiple active components in each 10 extract. Several active peaks are very selective for COX-2. Other Scutellaria sp.
including Scutellaria orthocalyx and Scutellaria lateriflora demonstrate a similar peak of inhibition (data not shown). However, both the COX-1 and COX-2 enzymes demonstrate multiple peaks of inhibition suggesting that there is more than one molecule contributing to the initial inhibition profiles.

Example 5. Isolation and Purification of the Active Free-B-ring Flavonoids from the Organic Extract of Scutellaria The organic extract (5 g) from the roots of Scutellaria orthocalyx, isolated as described in Example 1, was loaded onto prepacked flash column (120 g silica, 40 m particle size 32-60 m, 25 cm x 4 cm) and eluted with a gradient mobile phase of (A) 50:50 EtOAc:hexane and (B) methanol from 100% A to 100% B in 60 minutes at a flow rate of 15 mL/min. The fractions were collected in test tubes at 10 mL/fraction. The solvent was evaporated under vacuum and the sample in each fraction was dissolved in 1 mL of DMSO and an aliquot of 20 L was transferred to a 96 well shallow dish plate and tested for COX inhibitory activity. Based on the COX assay results, active fractions #31 to #39 were combined and evaporated. Analysis by HPLC/PDA and LC/MS showed a major compound with a retention times of 8.9 minutes and a MS peak at 272 m/e. The product was further purified on a C18 semi-preparation column (25 cm x 1 cm), with a gradient mobile phase of (A) water and (B) methanol, over a period of 45 minutes at a flow rate of 30. 5 mL/minute. Eighty-eight fractions were collected to yield 5.6 mg of light yellow solid.
Purity was determined by HPLC/PDA and LC/MS, and comparison with standards and NMR data. 1H NMR: S ppm. (DMSO-d6) 8.088 (2H, in, H-3',5'), 7.577 (3H, in, H-2',4',6'), 6.932 (1H, s, H-8), 6.613 (1H, s, H-3). MS: [M+1]+ = 271m/e. The compound was identified as baicalein. The IC50 of baicalein against the COX-2 enzyme was determined to be 10 g/mL.

Using preparative C-18 column chromatography, other Free-B-ring flavonoids were isolated and identified using a standardized extract isolated from the roots of Scutellaria baicalensis (lot # RM052302-01), having a Free-B-ring flavonoid content of 82.2%. Eleven structures were elucidated using HPLC/PDA/MS as illustrated in Figure 3.
With reference to Figure 3, the eleven compounds identified were baicalin, wogonin-7-glucuronide, oroxylin A 7-glucuronide, baicalein, wogonin, chrysin-7-glucuronide, 5-methyl-wogonin-7-glucuronide, scutellarin, norwogonin, chrysin and oroxylin A.

Example 6. COX Inhibition of Purified Free-B-ring Flavonoids Several Free-B-ring flavonoids have been obtained and tested at a concentration of 20 g/mL for COX-2 inhibitory activity using the methods described in Example 2. The results are summarized in Table 4.
Measurement of the IC50 of baicalein, baicalin and a standardized Free-B-ring flavonoid extract isolated from the roots of Scutellaria baicalensis was performed using the following method. A cleavable, peroxide chromophore was included in the assay to visualize the peroxidase activity of each enzyme in the presence of arachidonic acid as a cofactor. Typically, the assays were performed in a 96-well format. Each inhibitor, taken from a 10 mg/mL stock in 100% DMSO, was tested in triplicate at room temperature using the following range of concentrations: 0, 0.1, 1, 5, 10, 20, 50, 100, and 500 g/mL. To each well, 150 L of 100 mM Tris-HO, pH 7.5 was added along with 10 L of 22 M

Hematin diluted in tris buffer, 10 L of inhibitor diluted in DMSO, and 25 units of either COX-1 or COX-2 enzyme. The components were mixed for 10 seconds on a rotating platform, after which 20 L of 2 mM N,N,N'N'-Tetramethyl-p-phenylenediamine dihydrochloride (TMPD) and 20 gL of 1.1 mM AA was added to initiate the reaction. The plate was shaken for 10 seconds and then incubated for 5 minutes before reading the absorbance at 570 nm. The inhibitor concentration vs. percentage inhibition was plotted and the IC50 determined by taking the half-maximal point along the isotherm and intersecting the concentration on the x-axis. The IC50 was then normalized to the number of enzyme units in the assay. The dose response and IC50 results for baicalein, baicalin and a standardized Free-B-ring flavonoid extract isolated from the roots of Scutellaria baicalensis are provided in Figures 4, 5 and 6, respectively.

Table 4. Inhibition of COX Enzymatic Activity by Purified Free-B-ring Flavonoids Free-B-rin Flavonoids Inhibition of COX-1 Inhibition of COX-2 Baicalein 107% 109%
5,6-Dih drox -7-methoxyflavone 75% 59%
7,8-Dih droxyflavone 74% 63%
Baicalin 95 0 970!
Wogonin 16% 12%
Example 7. HPLC Quantification of Free-B-ring Flavonoids in Active Extracts Isolated from Scutellaria orthocalyx (roots), Scutellaria baicalensis (roots) and Oroxylum indicum (seeds) The presence and quantity of Free-B-ring flavonoids in five active extracts isolated from three different plant species have been confirmed and are set forth in the Table 5.
The Free-B-ring flavonoids were quantitatively analyzed by HPLC using a Luna C-column (250 x 4.5 mm, 5 m) a using 1% phosphoric acid and acetonitrile gradient from 80% to 20% in 22 minutes. The Free-B-ring flavonoids were detected using a W
detector at 254 nm and identified based on retention time by comparison with Free-B-ring flavonoid standards.

Table 5. Free-B-rin Flavonoid Content in Active Plant Extracts Weight of % Extractible Total amount % Free-B-ring Active Extracts Extract from BioMass of Free-B-ring Flavonoids in Flavonoids Extract S. orthocalyx 8.95 g 14.9% 0.2 mg 0.6%
(aqueous extract) S. orthocalyx 3.43 g 5.7% 1.95 mg 6.4%
(organic extract) S. baicalensis 7.18 g 12.0% 0.03 mg 0.07%
(aqueous extract) S. baicalensis 9.18 g 15.3% 20.3 mg 35.5%
(organic extract Oroxylum indicum 6.58 g 11.0% 0.4 mg 2.2%
(organic extract Example 8. Isolation and Purification of Active Compounds from the Organic Extract of Acacia catechu The organic extract (5 g) from the roots of A. catechu, isolated as described in Example 1, was loaded onto prepacked flash column (120 g silica, 40 m particle size 32-60 pm, 25 cm x 4 cm) and eluted with a gradient mobile phase of (A) 50:50 EtOAc:hexane and (B) methanol from 100% A to 100% B in 60 minutes at a flow rate of 15 mL/min.
The fractions were collected in test tubes at 10 mL/fraction. The solvent was evaporated under vacuum and the sample in each fraction was dissolved in DMSO (1 mL) and an aliquot of 20 L was transferred to a 96 well shallow dish plate and tested for COX
inhibitory activity. Based upon the COX assay results, active fractions #32 to #41 were combined and evaporated to yield 2.6 g of solid. Analysis by HPLC/PDA and LC/MS
showed two major compounds with retention times of 15.8 and 16.1 minutes, respectively.
The product was further purified on a C18 semi-preparatory column (25 cm x 1 cm), loaded with 212.4 mg of product and eluted with a gradient mobile phase of (A) water and (B) acetonitrile (ACN), over a period of 60 minutes at a flow rate of 5 mL/minute. Eighty-eight fractions were collected and two active compounds were isolated.
Compound 1 (11.5 mg) and Compound 2 (16.6 mg). Purity was determined by HPLC/PDA and LC/MS
data by comparison with standards (catechin and epicatechin) and NMR data.

Compound 1. 13C NMR: 8 ppm (DMSO-d6) 27.84 (C4), 66.27 (C3), 80.96 (C2), 93.78 (C9), 95.05 (C7), 99.00 (C5), 114.48 (C12), 115.01 (C15), 118.36 (C16), 130.55 (C11), 144.79 (C14), 155.31 (C6), 156.12 (CIO), 156.41 (C8). 1H NMR: 8 ppm.
(DMSO-d6) 9.150 (1H, s, OH), 8.911 (1H,s, OH), 8.835 (111, s, OH), 8.788 (111, s, OH), 6.706 (111, d, J=2 Hz, H2'), 6.670 (1H, d, J=8.0 Hz, H-6'), 6.578 (1H, dd, J=2, 8 Hz, H-5'), 5.873 (1H, d, J=2 Hz, H8), 5.670 (111, d, J=2 Hz, H6), 4.839 (1H, d, J=4 Hz, OH), 4.461 (111, d, J=7.3 Hz, H2), 3.798 (111, m, 113), 2.625 (111, m, H4b), 2.490 (111, m, H4a).
MS: [M+1]+
= 291 m/e. This compound was identified as catechin.

Compound 2. 13C NMR: S ppm. (DMSO-d6) 28.17 (C4), 64.87 (C3), 78.02 (C2), 94.03 (C9), 95.02 (C7), 98.44 (C5), 114.70 (C12), 114.85 (C15), 117.90 (C16), 130.56 (Cl l), 144.39 (C14), 155.72 (C6), 156.19 (CIO), 156.48 (C8). IH NMR: 8 ppm.
(DMSO-d6) 9.083 (1H, s, OH), 8.873 (1H,s, OH), 8.777 (1H, s, OH), 8.694 (1H, s, OH), 6.876 (1H, d, J=2 Hz, H2'), 6.646 (2H, s, H-5', 6'), 5.876 (1H, d, J=2 Hz, H8), 5.700 (1H, d, J=2 Hz, H6), 4.718 (1H, s, OH), 4.640 (1H, d, J=4.5 Hz, H2), 3.987 (1H, d, J=4.5 Hz, H3), 2.663 (1H, dd, J=4.6, 6.3 Hz, H4b), 2.463 (1H,dd, J=4.6, 6.3 Hz, H4a). MS:
[M+1]+
291 m/e. This compound was identified as epicatechin.

The dose response and IC50 results for catechin and a standardized flavan extract isolated from the bark of A. catechu are illustrated in Figures 7 and 8, using the method described in Example 6. The IC50 values of epicatechin against the COX-1 and enzymes are 7 g/mL and 20 g/mL, respectively.

Example 9. HPLC Quantification of Active Extracts from Acacia catechu The flavan content in the organic and aqueous extracts isolated from Acacia catechu were quantified by HPLC using a PhotoDiode Array detector (HPLC/PDA) and a Luna C18 column (250 mm x 4.6 mm). The flavans were eluted from the column using an acetonitrile gradient from 10% to 30% ACN over a period of 20 minutes, followed by 60%

ACN for five minutes. The results are set forth in Table 6. A profile of the HPLC
purification is shown in Figure 9. The flavans were quantified based on retention time and PDA data using catechin and epicatechin as standards. The retention times for the two major flavans were 12.73 minutes and 15.76 minutes, respectively.

Table 6. Free-B-ring Flavonoid Content in Active Plant Extracts Active Extracts from Weight of % Extractible % Flavans bark of A. catechu Extract from BioMass in Extract Aqueous Extract 10.8 g 18.0% 0.998%
Organic Extract 27.2 g 45.3% 30.37%

Example 10. In vitro Study of COX Inhibitory Activity of Organic Extracts from Acacia catechu and Scutellaria Species In vitro efficacy and COX-2 specificity of organic extracts isolated from Acacia catechu and various Scutellaria species were tested in cell-based systems for their ability to inhibit the generation of AA metabolites. Cell lines HOSC, which constitutively express COX-2 and TRIP-1, which express COX-1 were tested for their ability to generate PGE2 in the presence of AA.

COX-2 Cell Based Assay. HOSC (ATCC#8304-CRL) cells were cultured to 80-90% confluence. The cells were trypsinized, washed and resuspended in 10 ML at 1 x 106 cells/mL in tissue culture media (MEM). The cell suspension (200 AL) was plated out in 96-well tissue culture plates and incubated for 2 hours at 37 C and 5% CO2.
The media 5 was then replaced with new HOSC media containing 1 ng/mL IL-lb and incubated overnight. The media was removed again and replaced with 190 mL HOSC media.
Test compounds were then added in 10 L of HOSC media and were incubated for 15 minutes at 37 C. Arachidonic acid in HOSC media (20 mL, 100 AM) was added and the mixture was incubated for 10 minutes on a shaker at room temperature. Supernatant (20 AL) was 10 transferred to new plates containing 190 L/well of 100 M indomethacin in ELISA
buffer. The supernatants were analyzed as described below by ELISA.

COX-1 Cell Based Assay. THP-1 cells were suspended to a volume of 30 mL
(5x105 cells/mL). TPA was added to a final concentration of 10 nM TPA and cultured for 48 hours to differentiate cells to macrophage (adherent). The cells were resuspended in 15 HESS (25 mL) and added to 96-well plates in 200 mL volumes at 5 x 105 cells/well. The test compounds in RPMI 1640 (10 L) were then added and incubated for 15 minutes at 37 C. Arachidonic acid in RPMI (20 AL) was then added and the mixture was incubated for 10 minutes on a shaker at room temperature. Supernatant (20 AL) was added to ELISA
buffer (190 L) containing indomethacin (100 M). The supernatants were then analyzed 20 by ELISA, as described below.

COX-2 Whole Blood Assay. Peripheral blood from normal healthy donors was collected by venipuncture. Whole blood (500 L) was incubated with test compounds and extracts for 15 minutes at 37 C. Lipopolysaccharide (LPS, from E. coli serotype 0111:B4) was added to a final concentration of 100 ig/mL and cultured overnight at 37 C. The 25 blood was centrifuged (12,000 x g) and the plasma was collected. Plasma (100 AL) was added to methanol (400 L) to precipitate proteins. Supernatants were measured for PGE2 production by ELISA. This procedure is a modification of the methods described by Brideau et al. (1996) Inflamm. Res. 45:68-74.
COX-1 Whole Blood Assay. Fresh blood was collected in tubes not containing 30 anti-coagulants and immediately aliquoted into 500 L aliquots in siliconized microcentrifuge tubes. Test samples were added, vortexed and allowed to clot for 1 hour at 37 C. The samples were then centrifuged (12,000 x g) and the plasma was collected.
The plasma (100 AL) was added to methanol (400 AL) to precipitate proteins.
Supernatants were measured for TXB2 production by ELISA. This procedure is a modification of the methods described by Brideau et al. (1996) Inflamm. Res.
45:68-74.

ELISA Assays. Immunolon-4 ELISA plates were coated with capture antibody 0.5-4 pg/mL in carbonate buffer (pH 9.2) overnight at 4 C. The plates were washed and incubated for 2 hours with blocking buffer (PBS + 1% BSA) at room temperature.
The plates were washed again and test sample (100 L) was added and incubated for 1 hour at room temperature while shaking. Peroxidase conjugated secondary antibody was added in a 50 L volume containing 0.5-4 mg/mL and incubated for 1 hour at room temperature while shaking. The plates were then washed three times and TMB substrate (100 AL) was added. The plates were allowed to develop for 30 minutes, after which the reaction was stopped by the addition of 1 M phosphoric acid (100 L). The plates were then read at 450 run using a Wallas Victor 2 plate reader.

Cytotoxicity. Cellular cytotoxicity was assessed using a colorimetric kit (Oxford Biochemical Research) that measures the release of lactate dehydrogenase in damaged cells. Assays were completed according to manufacturer's directions. Both purified flavans and standardized extract from Acacia catechu were tested. No cytotoxicity was observed for any of the tested compounds.

The results of the assays are set forth in Table 7. The data are presented as values for direct comparison. With reference to Table 5, IC50 values are generally lower for COX-1 than COX-2. Additionally, whole blood was also measured for the differential inhibition of PGE2 generation (a measure of COX-2 in this system) or thromboxane B2 (TXB2) (a measure of COX-1 activation). Referring to Table 7, these studies clearly demonstrate specificity for COX-2 inhibition within the assays based on whole blood cells.
However, studies using the THP-1 and HOSC-based model system actually showed greater selectivity for COX-l. Possible reasons for this discrepancy are the fundamental differences between immortalized cell lines that constitutively express each of the enzymes and primary cells derived from whole blood that are induced to express COX
enzymes.
Primary cells are a more relevant model to study inflammation in vivo.
Additionally, the compounds used to identify COX-1 vs. COX-2 activity vary in each of these systems and consequently are not directly comparable.

Table 7 Inhibition of COX Activi in Cell Systems by Organic Extracts Plant Source of the Cell Line Based Assay Whole Blood Assay organic extracts IC50 COX-2 IC50 COX-1 IC50 COX-2 IC50 COX-1 A. catechu (bark) 78 g/mL 22 g/mL 40 mL >50 mL
S. orthocalyx (root) 50 mL 18 /ighnL 10 Ag/mL >50 Ag/mL
S. baicalensis (root) 82 mL 40 g/mL 20 mL 8 mL
S. lateriflora 60 Ag/mL 30 g/mL 8 Ag/mL 20 g/mL
(whole plant) Example 11. Inhibition of 5-Lipoxy eng ase by the Catechin from Acacia catechu As noted above, one of the most important pathways involved in the inflammatory response is produced by non-heme, iron-containing lipoxygenases (5-LO, 12-LO, and 15-LO), which catalyze the addition of molecular oxygen onto fatty acids such as AA (AA) to produce the hydroperoxides 5-, 12- and 15-BPETE, which are then converted to leukotrienes. There were early indications that the flavan extract from A.
catechu may provide some degree of 5-LO inhibition, thereby preventing the formation of 541PETE. A
Lipoxygenase Inhibitor Screening Assay Kit (Cayman Chemical, Inc., Cat#
760700) was used to assess whether the purified flavan catechin from Acacia catechu directly inhibited 5-LO in vitro. The 15-LO from soybeans normally used in the kit was replaced with potato 5-LO, after a buffer change from phosphate to a tris -based buffer using microfiltration was performed. This assay detects the formation of hydroperoxides through an oxygen sensing chromagen. Briefly, the assay was performed in triplicate by adding 90 L of 0.17 units/ L potato 5-LO, 20 L of 1.1 mM AA, 100 L of oxygen-sensing chromagen, and 10 L of purified flavan inhibitor to final concentrations ranging from 0 to 500 g/mL. The IC50 for 5-LO inhibition from catechin was determined to be 1.38 g/mL/unit of enzyme.

Example 12. Preparation of a Standardized Extract from Acacia catechu Acacia catechu (500 mg of ground bark) was extracted with the following solvent systems. (1) 100% water, (2) 80:20 water:methanol, (3) 60:40 water:methanol, (4) 40:60 water:methanol, (5) 20:80 water:methanol, (6) 100% methanol, (7) 80:20 methanol:THF, (8) 60:40 methanol:THF. The extracts were concentrated and dried under low vacuum.
The identification of the chemical components in each extract was achieved by HPLC
using a PhotoDiode Array detector (HPLC/PDA) and a 250 mm x 4.6 mm C18 column.
The chemical components were quantified based on retention time and PDA data using catechin and epicatechin as standards. The results are set forth in Table 8 and Figure 9.
As shown in Table 6, the flavan extract generated from solvent extraction with 80%
methanol/water provided the best concentration of flavan components.

Table 8. Solvents for Generating Standardized Flavan Extracts from Acacia catechu Extraction Weight of % Extractible Total amount of % Catechins Solvent Extract from BioMass Catechins in Extract 100% water 292.8 mg 58.56% 13 mg 12.02%
water:methanol 282.9 mg 56.58% 13 mg 11.19%
(80:20) water:methanol 287.6 mg 57.52% 15 mg 13.54%
(60:40) water:methanol 264.8 mg 52.96% 19 mg 13.70%
(40:60) water:methanol 222.8 mg 44.56% 15 mg 14.83%
(20:80) 100% methanol 215.0 mg 43.00% 15 mg 12.73%
methanol:THF 264.4 mg 52.88% 11 mg 8.81%
(80:20) methanol:THF 259.9 mg 51.98% 15 mg 9.05%
(60:40) Example 13. Preparation of Standardized Free-B-ring Flavonoid Extracts from various Scutellaria species Scutellaria orthocalyx (500 mg of ground root) was extracted twice with 25 mL
of the following solvent systems. (1) 100% water, (2) 80:20 water:methanol, (3) 60:40 water:methanol, (4) 40:60 water:methanol, (5) 20:80 water:methanol, (6) 100%
methanol, (7) 80:20 methanol:THF, (8) 60:40 methanol:THF. The extracts were combined, concentrated and dried under low vacuum. Identification of chemical components in each extract was performed by HPLC using a PhotoDiode Array detector (HPLC/PDA) and a 250 mm x 4.6 mm C18 column. The chemical components were quantified based on retention time and PDA data using baicalein, baicalin, scutellarein, and wogonin as standards. The results are set forth in Table 9.

Table 9. Quantification of Free-B-ring Flavonoids Extracted from Scutellaria orthocal x Extraction Weight of % Extractible Total amount % Flavonoids Solvent Extract from BioMass of Flavonoids in Extract 100% water 96 mg 19.2% 0.02 mg 0.20%
Water:methanol 138.3 mg 27.7% 0.38 mg 0.38%
(80:20) Water:methanol 169.5 mg 33.9% 0.78 mg 8.39%
(60:40) Water:methanol 142.2 mg 28.4% 1.14 mg 11.26%
(40:60) Water:methanol 104.5 mg 20.9% 0.94 mg 7.99%
(20:80) 100% methanol 57.5 m 11.5% 0.99 mg 10.42%
methanol:THF 59.6 mg 11.9% 0.89 mg 8.76%
(80:20) methanol:THF 58.8 mg 11.8% 1.10 mg 10.71%
(60:40) Scutellaria baicalensis (1000 mg of ground root) was extracted twice using 50 mL
of a mixture of methanol and water as follows: (1) 100% water, (2) 70:30 water:methanol, (3) 50:50 water:methanol, (4) 30:70 water:methanol, (5) 100% methanol. The extracts were combined, concentrated and dried under low vacuum. Identification of the chemical components was performed by IIPLC using a PhotoDiode Array detector (HPLC/PDA), and a 250 mm x 4.6 mm C18 column. The chemical components in each extract were quantified based on retention time and PDA data using baicalein, baicalin, scutellarein, and wogonin standards. The results are set forth in Table 10.
Table 10. Quantification of Free-B-ring Flavonoids Extracted from Scutellaria baicalensis Extraction Weight of % Extractible Total amount % Flavonoids Solvent Extract from BioMass of Flavonoids in Extract 100% water 277.5 mg 27.8% 1 mg 0.09%
Water:methanol 338.6 mg 33.9% 1.19 mg 11.48%
70:30 Water:methanol 304.3 mg 30.4% 1.99 mg 18.93%
(50:50) Water:methano1 293.9 mg 29.4% 2.29 mg 19.61%
(30:70) 100% methanol 204.2 m 20.4% 2.73 m 24.51%
Example 14. Preparation of a Formulation with a Standardized Free-B-ring Flavonoid Extract from the Roots of Scutellaria baicalensis and a Standardized Flavan Extract from the Bark of Acacia catechu 5 A novel composition of matter, referred to herein as UnivestinTM was formulated using two standardized extracts isolated from Acacia and Scutellaria, respectively, together with one or more excipients. A general example for preparing such a composition is set forth below. The Acacia extract used in this example contained >60%
total flavans, as catechin and epicatechin, and the Scutellaria extract contained >70% Free-B-ring 10 flavonoids, which was primarily baicalin. The Scutellaria extract contained other minor amounts of Free-B-ring flavonoids as set forth in Table 11. One or more exipients is added to the composition of matter. The ratio of flavan and Free-B-ring flavonoids can be adjusted based on the indications and the specific requirements with respect to inhibition of COX-2 vs. 5-LO and potency requirements of the product. The quantity of the 15 excipients can be adjusted based on the actual active content of each ingredient. A
blending table for each individual batch of product must be generated based on the product specification and QC results for individual batch of ingredients. Additional amounts of active ingredients in the range of 2-5% are recommended to meet the product specification. Table 11 illustrates a blending table that was generated for one batch of 20 UnivestinTM (Lot#G1702-COX-2).

Scutellaria baicalensis root extract (38.5 kg) (lot # RM052302-01) having a Free-B-ring flavonoid content of 82.2% (baicalin); Acacia catechu bark extract (6.9 kg) (lot #
RM052902-01) with total flavan content of 80.4%; and excipient (5.0 kg of Candex) were combined to provide a UnivestinTM formulation (50.4 kg) having a blending ratio of 85:15.
25 Table 9 provides the quantification of the active Free-B-ring flavonoids and flavans of this specific batch of UnivestinTM (Lot#G1702-COX-2), determined using the methods provided in Examples 7 and 9.

Table 11. Free-B-ring Flavonoid and Flavan Content of UnivestinTM Formulation Active Components % Content 1. Flavonoids a. Baicalin 62.5%
b. Minor Flavonoids i. Wo onin-7- lucuronide 6.7%
ii. Oroxylin A 7-glucuronide 2.0%
iii. Baicalein 1.5%
iv. Wogonin 1.1%
v. Chrysin-7-glucuronide 0.8%
vi. 5-Methyl-wogonin-7- lucuronide 0.5%
vii. Scuteliarin 0.3 o viii. Norwogonin 0.3%
ix. Chrysin <0.2%
x. Oroxylin A <0.2%

c. Total Free-B-ring Flavonoids 75.7%
2. Flavans a. Catechin 9.9%
b. Epicatechin 0.4%
c. Subtotal Flavans 10.3%
3. Total Active Ingredients 86%

With reference to Table 9, this specific batch of UnivestinTM contains 86%
total active ingredients, including 75.7% Free-B-ring flavonoids and 10.3% flavans.
Two different dosage levels of fmal product in capsule form were produced from this batch of UnivestinTM (50.0 kg): 125 mg per dose (60 capsules) and 250 mg per dose (60 capsules).
The final product was evaluated in a human clinical trial as described in Example 15.
Using the same approach, two other batches of UnivestinTM were prepared using a combination of a standardized Free-B-ring flavonoid extract from Scutellaria baicalensis roots and a standardized flavan extract from Acacia catechu bark having a blending ratio of 50:50 and 20:80, respectively.

Example 15. Measurements of Dose Response and ICso Values of COX Enzyme Inhibitions from Three Formulations of UnivestinTM
The three different formulations of UnivestinTM are produced as provided in Example 14 were tested for COX-1 and COX-2 inhibitory activity as described in Example 6. All three formulation show significant dose response inhibition of COX
enzyme activities as illustrated in Figures 11,12 and 13).

Exam le e 16. Measurements of Dose Response and IC50 Values of LO Enzyme Inhibition from a Formulation of UnivestinTm A Univestin sample was produced as outlined in Example 14, using a combination of a standardized Free-B-ring flavonoid extract from Scutellaria baicalensis roots and a standardized flavan extract from Acacia catechu bark with a blending ratio of 80:20. The sample was titrated in tissue culture media containing THP-l or HT-29 cells;

monocyte cell lines that express COX-1, COX-2 and 5-LO. A competitive ELISA
for LTB4 (LTB4; Neogen, Inc., Cat#4061 10) was used to assess the effect of UnivestinTM
on newly synthesized levels of LTB4 present in each cell line as a measure of UnivestinTM's inhibitory effect on the 5-LO pathway. The assay was performed in duplicate by adding 160,000 to 180,000 cells per well in 6-well plates. Univestin"' was added to the THP-1 cultures at 3, 10, 30 and 100 pg/mL and incubated overnight (-12-15 hrs) at 37 C with 5%
CO2 in a humidified environment. The results are set forth in Figure 14, which shows that the production of newly LPS-induced LTB4 was almost completely inhibited by the addition of Univestin to theTHP-1 cultures between 3 and 10 gg/mL.

Univestin and ibuprofen, another known 5-LO inhibitor, were added to the HT-29 cells at 3 g/mL and incubated 48 hrs at 37 C with 5% CO2 in a humidified environment. Each treated cell line was then harvested by centrifugation and disrupted by gentle dounce homogenization lysis in physiological buffers. As shown in Figure 15, Univestin inhibited generation of 80% of the newly synthesized LTB4 in HT-29 cells.
Ibuprofen only showed a 20% reduction in the amount of LTB4 over the same time period.

Example 17. Differential Inhibition of cox-2 but not cox-1 Gene Expression by Univestin'rm vs. Other NSAIDs To evaluate whether Univestin is operating on the genomic level, isolated human, peripheral blood monocytes (PBMCs) were stimulated with lipopolysaccharide (LPS), treated with UnivestinTM as illustrated in Example 14, celecoxib, ibuprofen or acetaminophen, and the total RNA produced was then harvested and evaluated by semi-quantitative RT-qPCR. Specifically, the assay was constructed by adding 130,000 cells per well in 6-well plates. The cells were then stimulated with 10 ng/mL LPS
and co-incubated with UnivestinTM at 1, 3, 10, 30 and 100 g/mL and celecoxib, ibuprofen and acetaminophen at 3 g/mL for 18 hours at 37 C with 5% CO2 in a humidified environment. Each cell-treatment condition was then harvested by centrifugation and total RNA produced was isolated using TRIzol reagent (InvitrogenTM Life Technologies, Cat#15596-026) and the recommended TRIzol reagent protocol. Total RNA was reverse transcribed using Moloney Murine Leukemia Virus reverse transcriptase (M-MLV
RT;
Promega Corp., Cat#M1701) using random hexamers (Promega Corp., Cat#C1181).

qPCR experiments were performed on an ABI Prism07700 Sequence Detection System using pre-developed validated Assays-on-Demand products (AOD from Applied Biosystems, Inc., Cat# 4331182) for 18S rRNA internal standard and gene specific assays.
Gene specific expression values were standardized to their respective 18S rRNA
gene expression values (internal control) and then the no-LPS no-drug treatment condition normalized to 100. Treatment conditions are relative to this null condition.

UnivestinTM decreased normalized gene expression of cox-2 by over 100-fold, while cox-1 normalized gene expression showed little variation. When PBMCs were treated with 3 g/mL of UnivestinTM, celecoxib, ibuprofen or acetaminophen, only UnivestinTM did not increase gene expression of cox-2. It is believed that this is the first report of changes in gene expression levels of eicosinoids, cytokines, chemokines and other genes implicated in pain and inflammation pathways following treatment with a mixture of Free-B-ring flavonoids and flavans using semi-quantitative RT-qPCR techniques. This work has been coupled work with ELISA-based assays to evaluate changes in protein levels as well as enzyme function assays to evaluate alterations in protein function. As a result of these studies, both genomic and proteomic coupled effects following treatment with UnivestinTM
have been demonstrated. Other studies cited in the literature have used protein specific methods to infer gene expression rather than show it directly. The results are set forth in Figures 16 and 17.

Example 18. Evaluation of the Efficacy of UnivestirP with in vivo Mouse Ear Swelling Model In order to test whether UnivestinTm could be used to treat inflammation in vivo, the composition, prepared as described in Example 14, was administered by oral gavage to 4-5 week old ICR mice (Harlan Labs) one day before treatment of their ears with AA.
Test mice were fed dose equivalents of 50, 100 and 200 mg/kg of Univestin suspended in olive oil while control mice were fed only olive oil. The following day, 20 L of 330 mM AA in 95% alcohol was applied to one ear of each mouse, while alcohol was applied to the other ear as a control. Mice treated with UnivestinTM showed a measurable dose response that tracked with increasing doses of UnivestinTM, as demonstrated in Figure 18.
With reference to Figure 18, the 200 mg/kg dose reduces swelling by over 50%
as compared to the minus UnivestinTM control. The 50 mg/kg dose of UnivestinTM
was as effective as the 50 mg/kg dose of another strong anti-inflammatory, indomethacin.
Example 19. Evaluation Efficacy of UnivestinTm with in vivo Mouse Ankle Joint Swelling Model Since UnivestinTM is designed to target joint pain, a solution of 20 L of 100 mM
AA in 95% ethanol was injected into the hind ankle joints of 4-5 week old ICR
mice (Harlan Labs) to generate swelling. The test group was fed 100 mg/kg of UnivestinTM
suspended in olive oil -12 hours before while another group was not given UnivestinTM
Control groups included mice that had not received arachidonic acid injections (negative control) and a group that had 95% ethanol without AA injected (vehicle control). These groups were also not given UnivestinTM. The results are set forth in Figure 19. With reference to Figure 19, the mice given UnivestinTM that were injected with AA
showed background levels of swelling as compared to the controls and the untreated arachidonic injected group. These results demonstrate the effectiveness of UnivestinTM for reducing swelling in joints, the site of action.

Example 20. Clinical Evaluation of the Efficacy of Free-B-ring Flavonoids and Flavans on the Relief of Pain Caused by Rheumatoid Arthritis or Osteoarthritis of the Knee and/or This clinical study was a single-center, randomized, double-blind, placebo-controlled study. Sixty subjects (n=60) with rheumatoid arthritis or osteoarthritis of the knee and/or hip were randomly placed into one of the following four groups:

5 A0 Placebo n=15 Placebo Ai Dose 1 n=15 UnivestinTM 250 mg/day (125 mg b.i.d.) A2 Dose 2 n=15 UnivestinTM 500 mg/day (250 mg b.i.d.) A3 Active Control n=15 Celecoxib 200 mg/day (100 mg b.i.d.) 10 The UnivestinTM was prepared as described in Example 14. This specific batch of Umvestm (lot#G1702-COX-2) contains 86% total active ingredients, including 11.5.11%
Free-B-ring flavonoids and 10.3% flavans. Celecoxib, also known as CelebrexTM, is a trade name for a prescription drug that is a COX-2 selective inhibitor.

Subjects were sex-matched and recruited from ages 40 to 75. Treatment consisted 15 of oral administration for 90 days of the placebo or active compound (UnivestinTM or celecoxib) according to the above dose schedule. Subjects taking NSAIDs engaged in a two-week washout period before beginning the study. Physical activity was not restricted, nor were the subjects given any advice as to diet. Subjects were free to withdraw from the trial at any time for any reason. The efficacy of the treatments was evaluated at 30, 60 and 20 90 days of oral administration by physicians, using the Western Ontario and McMaster Universities (WOMAC) Osteo-Arthritis Index (See Lingard et al. (2001) J. Bone & Joint Surg. 83:1856-1864; Soderman and Malchau (2000) Acta Orthop. Scand. 71 U139-46).
This protocol was reviewed and approved by an IRB board from University of Montreal.
The WOMAC was administered to subjects preferably in the doctor's office. They 25 were asked to read and respond to a questionnaire on their own or via proxy in the waiting room of the doctor's office or were interviewed by project personnel over the telephone and the data were transcribed in the computer database. This offered a stable environment among patients and reduced the possibility of bias due to different home environments among patients. Between groups differences for all measurements were evaluated with 30 One-Way Analysis of Variance and Tukey's Least Significant Difference for multiple comparisons. All questions were assigned a weight from 0 to 4 depending on the severity of pain, stiffness or impaired function. These values were then converted to percentages normalized to 100 and reported as WOMAC scores. Higher values are indicative of greater impairment. Table 12 sets forth the mean WOMAC index scores for pain, stiffness and function for 250 mg and 500 mg per day UnivestinTM compared to celecoxib at 200 mg per day and the placebo before treatment (baseline) and at 30, 60 and 90 days after treatment. The lower the score, the less pain and stiffness and better function a patient has.
Table 12. WOMAC Index Scores at Baseline and at 30, 60 and 90 Days UnivestinTm 250 Univestin~ 500 Celecoxib 200 Placebo WOMAC Mean Std Dev Mean Std Dev Mean Std Dev Mean Stu' Dev INDICE
Pain-baseline 54.33 19.9 60.33 23.34 55 22.28 49.33 15.1 Pain-30 days 41.33 19.22 36 22.93 50 23.09 41.67 15.55 Pain-60 days 40.71 16.62 40.77 19.77 30 16.46 57.31 16.66 Pain-90 days 41.79 16.36 27.69 21.57 31.67 16.42 50 14.43 Stiffness- 63.33 26.92 61.67 23.84 47.5 21.75 46.67 21.37 baseline Stiffness-30 41.67 16.14 44.17 21.06 39.42 18.29 59.17 20.85 days Stiffness-60 37.5 18.99 39.42 19.66 37.5 29.76 46.15 24.68 days Stiffness-90 39.29 20.72 28.85 21.28 29.17 25.19 49.04 18.01 days Function- 58.41 22.74 62.92 17.68 49.38 10.33 52.82 8.29 baseline Function-30 42.09 14.51 47.59 17.18 48.43 9.29 51.88 14.8 days Function-60 41.47 7.75 41.59 7.34 41.23 9.12 49.64 7.16 days Function-90 42.44 17.08 38.12 13.21 44.41 11.06 50.95 12.73 days Table 13 sets forth the mean absolute change in WOMAC scores for pain, stiffness and function. They are expressed as the difference between the baseline and the scores given at 30, 60 and 90 days. The more negative the score, the greater the improvement.

Table 13. Mean Absolute Change in WOMAC Scores at 30,60 and 90 Days*
Univestin1M 250 Univestin1M 500 Celecoxib 200 Placebo Absolute Mean Std Dev Mean Std Dev Mean Std Dev Mean Std Dev Change Pain-30 days -13 24.41 -24.33 18.7 -4.23 15.92 -7.67 26.98 Pain-60 days -14.64 26.85 -17.31 35.27 -22.31 22.51 5 13.54 Pain-90 days -13.57 22.91 -30.38 21.06 -16.67 21.36 -2.31 15.89 Stiffness-30 -21.67 24.31 -17.5 18.18 -8.65 20.66 12.5 29.88 days Stiffness-60 -28.57 27.05 -21.15 33.61 -9.62 29.82 -1.92 30.55 days Stiffness-90 -26.79 27.67 -31.73 20.17 -13.54 37.408 -0.96 26.74 days Function-30 -16.32 19.58 -15.33 18.28 -0.37 6.86 -0.94 14.05 days Function-60 -18.11 24.36 -21.4 19.79 -6.97 13.66 -3.49 11.81 days Function-90 -17.13 23.69 -24.87 23.25 -2.78 8.34 -2.18 11.27 days * These data contain only subjects who completed the study.
It is very difficult to ascribe a standard deviation to a group mean in a clinic trial due to the severe differences that appear in the data. Rather, confidence limits for the mean are preferred because they give a lower and upper limit for the mean and the narrower the interval, the more precise the estimate of the mean. Confidence limits are expressed in terms of a confidence coefficient. A 95% confidence interval is the most commonly used interval to describe a mean in this type of statistical analysis. This does not imply that there is a 95% probability that the interval contains the true mean. Instead, the level of confidence is associated with the method of calculating the interval. The confidence coefficient is simply the proportion of samples of a given size that may be expected to contain the true mean. That is, for a 95% confidence interval, if many samples are collected and the confidence interval computed, in the long run about 95%
of these intervals would contain the true mean. With this in mind, the 95% confidence interval was computed for the WOMAC scores for pain, stiffness and function at 30, 60 and 90 days.
Raw / non standardized scores for the WOMAC scores based on a five point Likert scale with a range between 1 and 5 were chosen to represent the final pain, stiffness and impaired function indices (Figures 20-31). Standardization to a scale between 0 and 100 was used in other sections for uniformity (see Tables 12 and 13) and to enhance the appreciation of the magnitudes of changes. However, given that all the figures are based on the same 1 - 5 point scales the raw data were plotted since they more accurately reflect the methods by which these scores were obtained from the patient questionnaires. In other words, since the patients were given a choice between 1 and 5 these representations better reflect the patient's response as opposed to the standardized or transformed score of 0 - 100 that does not reflect the patient's perception of possible range of answers.

Clear trends exist showing that for the pain indice that UnivestinTM at 250 and 500 mg/day reduced pain over the 90 day treatment period based on the patient responses.
Celecoxib also reduces pain over this same period of time compared to the placebo, which does not. However, celecoxib does not seem to be as effective as UnivestinTM
at both dosages in reducing stiffness, since the confidence intervals heavily overlapped those of the placebo. Finally, UnivestinTM at both doses clearly improved functional impairment, but celecoxib does not compared to placebo. The graphic representations contain all subjects even if they did not complete the study. Each confidence interval, however, is valid based on the number of subjects that were present at the time the WOMAC
tests were taken so the trends still hold. These data are plotted in Figures 20 through 31.
Example 21. Clinical Evaluation of the Efficacy of Free-B-ring Flavonoids and Flavans on BMI and Weight Loss Due to an Increase in Function.
Additional measurements taken during the clinical trial were height and weight.
All subjects in all groups (see Example 20) were measured for height and weight at 30 and 90 days of treatment. The subjects were given no recommendations on diet or exercise in order not to bias the results toward reduction of BMI and weight loss. Table 14 illustrates the changes in weight and BMI that occurred after treatment for 30 and 90 days.

Table 14. Change in Mean Weight (kg) and BMI (kg/m2) at 30 and 90 Days Group Univestinn 250 Univestinn 500 Celecoxib 200 Placebo Std Std Std Std Mean Dev Mean Dev Mean Dev Mean Dev eight -30 days -3.60 3.76 -2.40 3.31 -2.00 3.08 -.60 1.99 Weight -90 days -5.36 3.43 -4.15 4.81 -3.17 4.88 -.08 1.50 3MI -30 days -1.28 1.33 -.80 1.13 -.68 1.06 -.20 .64 3MI-90 days -1.84 1.14 -1.39 1.64 -1.07 1.67 -.02 .54 Based on these data, the 250 mg/day dose of UnivestinTM gave the greatest amount of weight loss and change in BMI followed by the 500 mg/day dose of UnivestinTM and then celecoxib. The placebo had no effect on weight or BMI.
It is not believed that there are any other reports in the literature of anti-inflammatory compounds being used to effect weight loss or changes in BMI.
Though the subjects were given no advice on exercise, the greater functional capabilities gained after treatment, especially with UnivestinTM, may have allowed them to exercise more on their own accord. Alternatively, UnivestinTM may be increasing thermogenesis, lipolysis, or causing an under utilization of carbohydrates or fat in the diet. Figures 32 and 33 illustrate the BMI and weight loss seen for UnivestinTM after 30 and 90 days of treatment.

Example 22. Clinical Evaluation of the Efficacy of Free-B-ringFlavonoids and Flavans on Lowering of Blood Glucose Due to an Increase in Function.
Blood glucose was also taken at 0 (baseline), 30 days and 90 days after treatment (see Example 20). These measurements were reported in mmole per liter. The data is also shown in mg/dL. Table 15 sets forth blood glucose levels after 30 and 90 days of treatment with UnivestinTM at 250 and 500 mg/day.

Table 15. Change in Blood Glucose after 30 and 90 days of Treatment Group Univestin M 250 Univestin M 500 Placebo Std Std Std mmoll mg/dL Dev mmol/L mg/dL Dev mmol/L mg/dL Dev L
Glucose- 5.24 94.32 .74 5.09 91.62 .67 4.82 86.76 .80 Baseline Glucose- 5.10 91.80 .71 4.75 85.50 .55 5.08 91.44 .54 30 days Glucose- 4.88 87.84 .72 4034 78.12 .36 4.71 84.78 .56 90 days Percent Change -7.52 -12.79 .94 By 90 days These data suggest that both the 250 and the 500 mg/day doses of UnivestinTM
are significantly lowering blood glucose levels over time. This impact may or may not be 5 related to the loss of weight observed above or to the presumed increase in activity as functional impairment improved. It may also be possible that UnivestinTM is acting directly to improve glucose metabolism by decreasing glucose tolerance or by utilizing carbohydrates more effectively.

Claims (11)

1. A composition of matter comprised of a comprised of a mixture of at least one Free-B-ring flavonoid and at least one flavan.
2. The composition of claim 1 wherein the ratio of Free-B-ring flavonoid to flavan in said composition is selected from the range of 99:1 Free-B-ring flavonoid:flavan to 1:99 of Free-B-ring flavonoid:flavan.
3. The composition of claim 2 wherein the ratio of Free-B-ring flavonoid:flavan in the composition of matter is about 85:15.
4. The composition of claim 1 wherein said Free-B-ring flavonoid is selected from the group of compounds having the following structure:

wherein R1, R2, R3, R4, and R5 are independently selected from the group consisting of -H, -OH, -SH, -OR, -SR, -NH2, -NHR, -NR2, -NR3+X-, a carbon, oxygen, nitrogen or sulfur, glycoside of a single or a combination of multiple sugars including, aldopentoses, methyl-aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof;
wherein R is an alkyl group having between 1-10 carbon atoms; and X is selected from the group of pharmaceutically acceptable counter anions including, hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride and carbonate.
5. The composition of claim 1 wherein said flavan is selected from the group of compounds having the following structure:

wherein R1, R2, R3, R4 and R5 are independently selected from the group consisting of H, -OH, -SH, -OCH3, -SCH3, -OR, -SR, -NH2, -NRH, -NR2, -NR3+X-, esters of substitution groups, independently selected from the group consisting of gallate, acetate, cinnamoyl and hydroxyl-cinnamoyl esters, trihydroxybenzoyl esters and caffeoyl esters; a carbon, oxygen, nitrogen or sulfur glycoside of a single or a combination of multiple sugars including, aldopentoses, methyl aldopentose, aldohexoses, ketohexose and their chemical derivatives thereof; dimer, trimer and other polymerized flavans;
wherein R is an alkyl group having between 1-10 carbon atoms ; and X is selected from the group of pharmaceutically acceptable counter anions including, but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride, carbonate.
6. The composition of claim 1 wherein said Free-B-ring flavonoid and said flavan are obtained by organic synthesis or are isolated from a plant.
7. The composition of claim 6 wherein said Free-B-ring flavonoid and said flavan are isolated from a plant part selected from the group consisting of stems, stem barks, trunks, trunk barks, twigs, tubers, roots, root barks, young shoots, seeds, rhizomes, flowers and other reproductive organs, leaves and other aerial parts.
8. The composition of claim 6 wherein said Free-B-ring flavonoid is isolated from a plant family selected from the group consisting of Annonaceae, Asteraceae, Bignoniaceae, Combretaceae, Compositae, Euphorbiaceae, Labiatae, Lauranceae, Leguininosae, Moraceae, Pinaceae, Pteridaceae, Sinopteridaceae, Ulmaceae and Zingiberacea.
9. The composition of claim 6 wherein said Free-B-ring flavonoid is isolated from a plant genus selected from the group consisting of Desmos, Achyrocline, Oroxylum, Buchenavia, Anaphalis, Cotula, Gnaphalium, Helichrysum, Centaurea, Eupatorium, Baccharis, Sapium, Scutellaria, Molsa, Colebrookea, Stachys, Origanum, Ziziphora, Lindera, Actinodaphne, Acacia, Derris, Glycyrrhiza, Millettia, Pongamia, Tephrosia, Artocarpus, Ficus, Pityrogramma, Notholaena, Pinus, Ulmus and Alpinia.
10. The composition claim 6 wherein said flavan is are isolated from a plant species selected from the group consisting of the Acacia catechu, Acacia concinna, Acacia farnesiana, Acacia Senegal, Acacia speciosa, Acacia arabica, A. caesia, A.
pennata, A.
sinuata. A. mearnsii, A. picnantha, A. dealbata, A. auriculiformis, A.
holoserecia and A.
mangium.
11. The composition of claim 6 wherein said Free-B-ring flavonoid is isolated from a plant or plants in the Scutellaria genus of plants and said flavan is isolated from a plant or plants in the Acacia genus of plants.
CA2703698A 2002-04-30 2003-04-30 Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent Expired - Lifetime CA2703698C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US37716802P 2002-04-30 2002-04-30
US60/377,168 2002-04-30
CA2484192A CA2484192C (en) 2002-04-30 2003-04-30 Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA2484192A Division CA2484192C (en) 2002-04-30 2003-04-30 Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent

Publications (2)

Publication Number Publication Date
CA2703698A1 true CA2703698A1 (en) 2003-11-13
CA2703698C CA2703698C (en) 2016-11-22

Family

ID=29401452

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2484192A Expired - Lifetime CA2484192C (en) 2002-04-30 2003-04-30 Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent
CA2703698A Expired - Lifetime CA2703698C (en) 2002-04-30 2003-04-30 Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CA2484192A Expired - Lifetime CA2484192C (en) 2002-04-30 2003-04-30 Formulation of a mixture of free-b-ring flavonoids and flavans as a therapeutic agent

Country Status (16)

Country Link
US (5) US7514469B2 (en)
EP (2) EP2108370A1 (en)
JP (2) JP4723239B2 (en)
KR (1) KR100621234B1 (en)
CN (2) CN101837003B (en)
AT (1) ATE438393T1 (en)
AU (1) AU2003228777C1 (en)
BR (1) BRPI0309689B1 (en)
CA (2) CA2484192C (en)
DE (1) DE60328676D1 (en)
DK (1) DK1503778T3 (en)
ES (1) ES2330097T3 (en)
HK (2) HK1073247A1 (en)
NZ (1) NZ535988A (en)
RU (1) RU2379031C2 (en)
WO (1) WO2003092599A2 (en)

Families Citing this family (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7108868B2 (en) 2002-03-22 2006-09-19 Unigen Pharmaceuticals, Inc. Isolation of a dual cox-2 and 5-lipoxygenase inhibitor from acacia
US20050096281A1 (en) * 2002-03-01 2005-05-05 Unigen Pharmaceuticals, Inc. Formulation of a mixture of Free-B-Ring flavonoids and flavans for use in the prevention and treatment of cognitive decline and age-related memory impairments
US7972632B2 (en) 2003-02-28 2011-07-05 Unigen Pharmaceuticals, Inc. Identification of Free-B-Ring flavonoids as potent COX-2 inhibitors
US8945518B2 (en) 2002-04-30 2015-02-03 Unigen, Inc. Formulation of dual eicosanoid system and cytokine system inhibitors for use in the prevention and treatment of oral diseases and conditions
US8034387B2 (en) 2002-04-30 2011-10-11 Unigen, Inc. Formulation of a mixture of free-B-ring flavonoids and flavans for use in the prevention and treatment of cognitive decline and age-related memory impairments
JP4723239B2 (en) 2002-04-30 2011-07-13 ユニジェン・インコーポレーテッド Formulation of a mixture of free-B-ring flavonoids and flavans as therapeutic agents
US20040097429A1 (en) * 2002-11-18 2004-05-20 Nieuwenhuizen Arie Gijsbert Method for the reduction of the mammalian appetite
JP2006519231A (en) * 2003-02-26 2006-08-24 ユニゲン・ファーマシューティカルス・インコーポレーテッド Formulations for the prevention and treatment of carbohydrate-induced diseases and conditions
CN1798568B (en) * 2003-04-04 2011-05-11 尤尼根制药公司 Formulation of dual cycloxygenase (cox) and lipoxygenase (lox) inhibitors for mammal skin care
EP1653914A4 (en) 2003-08-12 2008-10-29 3M Innovative Properties Co Oxime substituted imidazo-containing compounds
US7897597B2 (en) 2003-08-27 2011-03-01 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
WO2005023190A2 (en) 2003-09-05 2005-03-17 3M Innovative Properties Company Treatment for cd5+ b cell lymphoma
BRPI0414856A (en) 2003-10-03 2006-11-21 3M Innovative Properties Co alkoxy-substituted imidazoquinolines
US7544697B2 (en) 2003-10-03 2009-06-09 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and analogs thereof
AU2004291101A1 (en) 2003-11-14 2005-06-02 3M Innovative Properties Company Oxime substituted imidazo ring compounds
EP1682544A4 (en) 2003-11-14 2009-05-06 3M Innovative Properties Co Hydroxylamine substituted imidazo ring compounds
BRPI0416936A (en) 2003-11-25 2007-01-16 3M Innovative Properties Co substituted imidazo ring systems and methods
US8802853B2 (en) 2003-12-29 2014-08-12 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
AU2004312508A1 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Imidazoquinolinyl, imidazopyridinyl, and imidazonaphthyridinyl sulfonamides
TW200612932A (en) 2004-03-24 2006-05-01 3M Innovative Properties Co Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
BRPI0511227A (en) 2004-06-04 2007-11-27 Horizon Science Pty Ltd molasses extract, sugar sludge extract, broth and / or foam extract collected from the clarification tank, extract of fibrous / residual sugarcane stalks from the sugar beet or sugarcane field, bagasse extract and extract of sugar beet pulp having characteristics reducing ig or burning rate; method for the extraction of non-nutrient phytochemicals which have reducing properties of ig or the rate of burning of sugar processing waste stream and other products in process and sugarcane milling sludge; method for decreasing the ig of a food product; sucrose product containing low ig; method for obtaining food products having a lower ig or lower burn rate; method to improve health; method for decreasing the ig of a sucrose-containing product; low ig product; purified phytochemical extracted from sugar cane or sugar beet; low ig sweetener and sweetener
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
WO2006065280A2 (en) 2004-06-18 2006-06-22 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and methods
WO2006038923A2 (en) 2004-06-18 2006-04-13 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
KR100610562B1 (en) * 2004-06-28 2006-08-08 재단법인서울대학교산학협력재단 Compositions for preventing or treating an acute or chronic neurodegenerative diseases comprising flavonoide derivatives
KR100545304B1 (en) * 2004-09-01 2006-05-08 주식회사 유니젠 Composition comprising uncaria genus plant having uncaria gambir, or scutellaria radix and/or green tea extract mixture for suppressing cyclooxygenase or 5-lipoxygenase
US7691418B2 (en) * 2004-09-17 2010-04-06 Oystershell Nv Composition for inhibiting or preventing the formation of a biofilm
TW200630102A (en) * 2004-10-19 2006-09-01 Unigen Pharmaceuticals Inc Formulation of dual eicosanoid system and cytokine system inhibitors for use in the prevention and treatment of oral diseases and conditions
US20060140881A1 (en) 2004-12-22 2006-06-29 Guofeng Xu Oral care compositions containing flavonoids and flavans
CN101141999A (en) * 2004-12-22 2008-03-12 高露洁-棕榄公司 Oral care compositions containing free-b-ring flavonoids andflavans
WO2006074003A2 (en) 2004-12-30 2006-07-13 3M Innovative Properties Company CHIRAL FUSED [1,2]IMIDAZO[4,5-c] RING COMPOUNDS
JP5313502B2 (en) 2004-12-30 2013-10-09 スリーエム イノベイティブ プロパティズ カンパニー Substituted chiral condensed [1,2] imidazo [4,5-c] cyclic compounds
CA2597092A1 (en) 2005-02-04 2006-08-10 Coley Pharmaceutical Group, Inc. Aqueous gel formulations containing immune reponse modifiers
WO2006086634A2 (en) 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
JP4823538B2 (en) * 2005-02-28 2011-11-24 株式会社mimozax Anti-obesity composition containing acacia bark
RU2411941C2 (en) * 2005-03-10 2011-02-20 Юниджен Фармасьютикалз, Инк. Preparation based on mixture of flavonoids with free b-cycle and flavanes, as therapeutic agent
EP1863814A1 (en) 2005-04-01 2007-12-12 Coley Pharmaceutical Group, Inc. 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
EP2450084B1 (en) 2005-06-03 2014-02-19 Horizon Science Pty Ltd Substances having body mass redistribution properties
KR100729731B1 (en) 2005-07-19 2007-06-20 건국대학교 산학협력단 3,4'-dihydroxyflavone having activity of suppressing apoptosis of keratinocyte
AU2006308596A1 (en) * 2005-11-01 2007-05-10 Mars, Incorporated Flavanols and B-type procyanidins and inflammation
US20070154576A1 (en) * 2005-12-09 2007-07-05 Tripp Matthew L Protein kinase modulation by hops and Acacia products
US20070140990A1 (en) * 2005-12-21 2007-06-21 Nataly Fetissova Oral Compositions Comprising Propolis
JPWO2007108042A1 (en) * 2006-03-15 2009-07-30 株式会社トロピカルテクノセンター Anti-inflammatory agent
US20080003300A1 (en) * 2006-06-30 2008-01-03 Gaffar Maria C Compostions containing Mg/Zn/F-CaP plus inhibitors of pro-inflammatory Cytokines (a combination of a Free-B-Ring flavonoids and a flavan) for osteoporosis prevention, therapy and treatment of bone diseases
US7906506B2 (en) 2006-07-12 2011-03-15 3M Innovative Properties Company Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US9682256B2 (en) * 2006-07-14 2017-06-20 Colgate-Palmolive Company Methods of making compositions comprising films
AU2006347125B2 (en) * 2006-08-10 2013-06-06 Acacia-No-Ki Co., Ltd. Antioxidant composition containing component originating in the bark of tree belonging to the genus Acacia
KR101398737B1 (en) * 2006-08-10 2014-05-27 가부시키가이샤 미모잭스 Composition for preventing and/or treating tumor containing component originating in the bark of tree belonging to the genus acacia
AU2006347122B2 (en) * 2006-08-10 2013-06-06 Acacia-No-Ki Co., Ltd. Antiobesity composition containing component originating in the bark of tree belonging to the genus Acacia
WO2008018141A1 (en) * 2006-08-10 2008-02-14 Wood One Co., Ltd. Composition for preventing and/or treating itching containing component originating in the bark of tree belonging to the genus acacia
EP2052728B1 (en) * 2006-08-10 2018-06-13 mimozax Co., Ltd. Hypoglycemic composition containing component originating in the bark of tree belonging to the genus acacia
CN101553582A (en) * 2006-09-19 2009-10-07 视界科技有限公司 Extracts derived from sugar cane and a process for their manufacture
KR100761248B1 (en) * 2006-10-12 2007-10-04 주식회사 유니젠 Composition for treating atopic dermatitis comprising extracts of bamboo and scutellaria
US8517728B2 (en) * 2007-01-24 2013-08-27 Colgate-Palmolive Company Oral care implement having fluid delivery system
WO2008131119A2 (en) * 2007-04-17 2008-10-30 Acropolis Pharmaceuticals, Inc. Composition and method for cancer treatment and prevention
CN104644468A (en) * 2008-02-08 2015-05-27 高露洁-棕榄公司 Oral care product and methods of use thereof
EP2247290A1 (en) * 2008-02-25 2010-11-10 Nestec S.A. Polyphenols for the treatment of cartilage disorders
TWI366466B (en) * 2008-05-08 2012-06-21 Ind Tech Res Inst Herbal extracts capable of inducing immune cells to produce interferon and activating toll-like receptors and the preparation thereof
AU2010206549B2 (en) * 2009-01-26 2016-08-25 Dcb-Usa, Llc Use of pterosin compounds for treating diabetes and obesity
CN106822080A (en) 2009-04-29 2017-06-13 阿马里纳药物爱尔兰有限公司 Pharmaceutical composition containing EPA and cardiovascular agents and use its method
KR101582907B1 (en) * 2010-10-27 2016-01-08 주식회사 성균바이오텍 Composition comprising extract of Eupatorium spp. as an effective component for prevention or treatment of obesity or metabolic bone disease
CA2860448C (en) 2011-02-08 2019-09-10 Horizon Science Pty Ltd Molasses extract enriched in polyphenols and uses thereof
CA2829119A1 (en) 2011-03-09 2012-09-13 Colgate-Palmolive Company Interdental cleaning device
KR20120132114A (en) * 2011-05-27 2012-12-05 한국생명공학연구원 Pharmaceutical composition for the prevention or treatment of asthma containing wogonin as an effective ingredient
JP6203469B2 (en) * 2011-06-03 2017-09-27 花王株式会社 Dentifrice composition
CN102579590A (en) * 2012-02-18 2012-07-18 贾古友 Drug for treating knee osteoarthritis (OA)
CN104270945B (en) 2012-03-19 2017-03-29 巴克老龄化研究所 APP specific b ACE inhibitor (ASBI) and application thereof
CN102755311A (en) * 2012-07-05 2012-10-31 吉林大学 Application of oroxylin A in preparing medicament for treating pneumonia
JP6239622B2 (en) 2012-08-28 2017-11-29 ザ プロダクト メーカーズ (オーストラリア) プロプライエタリー リミテッド Extraction method
US9171343B1 (en) 2012-09-11 2015-10-27 Aseko, Inc. Means and method for improved glycemic control for diabetic patients
US9897565B1 (en) 2012-09-11 2018-02-20 Aseko, Inc. System and method for optimizing insulin dosages for diabetic subjects
IN2015DN03043A (en) 2012-10-26 2015-10-02 Colgate Palmolive Co
CN105452269B (en) 2013-06-17 2020-01-10 尤妮金公司 Compositions and methods for joint health
BR112016002244A2 (en) * 2013-08-02 2017-08-01 Council Scient Ind Res ulmosid-a: useful for the prevention or cure of metabolic diseases
EP3035949B1 (en) 2013-08-16 2023-10-04 Poly Gain Pte Ltd Sugar cane derived extracts and uses
US9898585B2 (en) 2014-01-31 2018-02-20 Aseko, Inc. Method and system for insulin management
US9486580B2 (en) 2014-01-31 2016-11-08 Aseko, Inc. Insulin management
US10583161B2 (en) * 2014-06-16 2020-03-10 Unigen, Inc. Compositions and methods for managing or improving bone disorders, joint disorders, cartilage disorders, or a combination thereof
CN107206040A (en) * 2014-06-16 2017-09-26 尤尼根公司 For managing or improving bone disorders, cartilage disorder or combination thing and method
CN104083390A (en) * 2014-06-28 2014-10-08 广西大学 Application of 6,7-dyhydroxyl-5-beta-D-glucopyranose-4'-beta-O-D-glucopyranose flavonoid in preparing anti-inflammation medicaments
US11081226B2 (en) 2014-10-27 2021-08-03 Aseko, Inc. Method and controller for administering recommended insulin dosages to a patient
EP3050023B1 (en) 2014-10-27 2021-08-25 Aseko, Inc. Subcutaneous outpatient management
CN104721210B (en) * 2015-03-16 2017-10-13 中国药科大学 Application of the oroxylin glycosides in medicine for treating tumor metastasis is prepared
WO2017031440A1 (en) 2015-08-20 2017-02-23 Aseko, Inc. Diabetes management therapy advisor
CN106727492A (en) * 2016-12-22 2017-05-31 黄冈师范学院 A kind of application of polymethoxyflavone on the medicine for preparing treatment cutaneous papilloma
EP3638277A4 (en) * 2017-06-15 2021-03-17 Sami Labs Limited Compositions and methods for beta secretase inhibition
US20210052543A1 (en) * 2018-01-09 2021-02-25 Brigham Young University Compositions and Methods for Treating Pain with Wogonin
CN108610394B (en) * 2018-05-07 2021-08-31 深圳市维琪医药研发有限公司 Preparation and purification method and application of peptidomimetic compound
CN114617968B (en) 2019-04-09 2023-10-03 北京五和博澳药业股份有限公司 Flavone polyphenol medicine self-emulsifying composition, preparation method thereof, medicine composition and application
JP2022550671A (en) * 2019-08-26 2022-12-05 サミ-サビンサ グループ リミテッド Plant active substances and their anti-pollution effect
CN117120043A (en) * 2020-07-30 2023-11-24 尤尼根公司 Standardized bioflavonoid compositions for regulating host defense mechanism homeostasis
CN112156120B (en) * 2020-11-09 2021-11-02 中国科学院新疆理化技术研究所 Application of Turkey jasmine leaf n-butyl alcohol extract
DE202022106782U1 (en) 2022-12-03 2022-12-19 Mohammed Azeemuddin Antiurolithiatic, antiosteoporotic, antidiabetic and anti-inflammatory composition of a polyherbal extract

Family Cites Families (205)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3686872A (en) * 1970-06-30 1972-08-29 Anthony James Whitworth Soil grouting process
US3706581A (en) * 1970-10-05 1972-12-19 Borden Co Soil grouting process and composition
US4035510A (en) * 1976-09-15 1977-07-12 Scott Eugene J Van Treatment of acne utilizing N-methyldiethanolamine
US4201776A (en) * 1977-01-31 1980-05-06 The Green Cross Corporation Food additive for reinforcing foods deficient in fiber content
GB2024817B (en) 1978-03-15 1982-09-22 Wellcome Found Flavan derivatives
US4268517A (en) * 1979-08-30 1981-05-19 Continental Pharma Pharmaceutical composition and therapeutical method for treating degenerative affections of the articular cartilage
JPS5738721A (en) 1980-08-20 1982-03-03 Wataru Sakai Production of bamboo extract having effect for hemorrhoids
JPS57109720A (en) * 1980-12-22 1982-07-08 Continentalfuaama Medicine containing (+)-catechine and/or its salt for degenerated disease of articular gristle
US4374824A (en) * 1981-01-27 1983-02-22 Krishan Dyal Mathur Dentifrice
GB2115409B (en) * 1982-02-24 1985-08-07 Zyma Sa Novel crystal modifications of (+)-catechin
JPS6150921A (en) 1984-08-20 1986-03-13 Ichimaru Fuarukosu Kk Purification of baicalin
JPS6183179A (en) 1984-09-28 1986-04-26 Otsuka Pharmaceut Co Ltd Novel flavone derivative
JPS61161219A (en) 1985-01-08 1986-07-21 Osaka Chem Lab Skin composition for atopic dermatitis
JPS61233627A (en) 1985-04-10 1986-10-17 Agency Of Ind Science & Technol Agent for suppressing degranulation of mastocyte
JPS61238719A (en) 1985-04-17 1986-10-24 Shiseido Co Ltd Hair cosmetic
JPH0755895B2 (en) 1985-08-29 1995-06-14 サンスター株式会社 Toothpaste composition
US4627977A (en) 1985-09-13 1986-12-09 Colgate-Palmolive Company Anticalculus oral composition
JPS6327435A (en) 1986-07-18 1988-02-05 Ichimaru Pharcos Co Ltd Extracting method for extract with high baicalein content
AU7873187A (en) * 1986-08-08 1988-02-24 University Of Minnesota Method of culturing leukocytes
IN168400B (en) * 1987-01-30 1991-03-23 Colgate Palmolive Co
US5156835A (en) * 1987-01-30 1992-10-20 Colgate-Palmolive Company Antibacterial antiplaque oral composition
CA1308661C (en) 1987-06-26 1992-10-13 Masayasu Kurono AGENT FOR INHIBITING BINDING OF 5.alpha.-DIHYDRO-TESTOSTERONE WITH ANDROGEN RECEPTOR AS WELL AS PROCESS FOR OBTAINING SAME
US5098709A (en) * 1987-07-21 1992-03-24 Kang Kwon J Method of administrating a fused salt from natural substances, namely ginkgo, persimmon, pine, and bamboo in the treatment of inflammations
JPH0662408B2 (en) 1987-10-01 1994-08-17 太陽化学株式会社 Anti-caries and anti-periodontal composition
FR2628317B1 (en) * 1988-03-09 1991-11-08 Lvmh Rech COMPOSITION BASED ON HYDRATED LIPID LAMID PHASES OR LIPOSOMES CONTAINING SCUTELLARIA EXTRACT, OR AT LEAST ONE FLAVONOID SUCH AS BAICALINE OR BAICALINE AND COSMETIC OR PHARMACEUTICAL COMPOSITION, IN PARTICULAR DERMATOLOGICAL, ANTI-ALLERGIC ANTI-ALLERGIC ACTIVITY THE INCORPORANT
US4965067A (en) * 1988-08-10 1990-10-23 The Proctor & Gamble Company Oral compositions
US4942333A (en) 1988-12-05 1990-07-17 North American Philips Corporation Shadow mask with border pattern
JPH0755895Y2 (en) 1989-04-04 1995-12-25 函館製網船具株式会社 Multi-stage aquaculture cage
US4946684A (en) * 1989-06-20 1990-08-07 American Home Products Corporation Fast dissolving dosage forms
FR2651132B1 (en) * 1989-08-30 1993-01-08 Pacific Chem Co Ltd PROTECTIVE AGENTS FOR CELLS AGAINST CHEMICAL SPECIES WITH ACTIVE OXYGEN AND THEIR PREPARATION.
JPH03240725A (en) * 1990-02-15 1991-10-28 Senjiyu Seiyaku Kk Maillard reaction inhibitor
JPH03251518A (en) 1990-02-26 1991-11-11 Ichimaru Pharcos Co Ltd Skin, scalp and hair agent containing component of suppressing growth of dandruff fungus extracted from bamboo
JP3133095B2 (en) 1990-04-25 2001-02-05 兆岐 史 Gastrointestinal sclerosis agent
JP2704783B2 (en) 1990-05-23 1998-01-26 長谷川香料株式会社 Prevention method of flavor deterioration of food and drink
CN1028604C (en) 1990-06-13 1995-05-31 成都中医学院 Prepn. of Chinese herb-fresh bamboo-juice
US5585371A (en) 1990-08-29 1996-12-17 Humanetics Corporation Treatment of immune system with Δ5-androstenes
US5096701A (en) * 1990-12-18 1992-03-17 The Procter & Gamble Company Oral compositions
JP3802555B2 (en) * 1991-12-17 2006-07-26 フイズ テクノロジーズ リミテッド Ulcer prevention and treatment composition and method
JPH05271088A (en) 1991-12-27 1993-10-19 Ruibosuteii Japan:Kk Agent for improving and treating tissue disorder caused by dermatosis-immunological reaction
FR2687572B1 (en) 1992-02-26 1995-06-09 Oreal USE OF CERTAIN FLAVONOUIDS IN COSMETIC COMPOSITIONS, AND METHOD FOR PROTECTING THE SKIN, ITS PHANERES AND COSMETIC COMPOSITIONS.
JPH05331061A (en) 1992-06-01 1993-12-14 Tsumura & Co Apoptosis-inducing agent
JPH05331161A (en) 1992-05-27 1993-12-14 Tsumura & Co Chromone derivative and aldose reductase inhibitor containing the compound as active component
US5605929A (en) 1992-05-27 1997-02-25 Arch Development Corp. Methods and compositions for inhibiting 5α-reductase activity
JPH08104268A (en) 1992-07-23 1996-04-23 Joji Maruyama Parking device for bicycle
US5650432A (en) * 1995-03-24 1997-07-22 Jlb, Inc. Method of treating or preventing non-viral microbial infection
US5320830A (en) * 1992-12-30 1994-06-14 The Procter & Gamble Company Oral compositions
US5364845C1 (en) 1993-03-31 2002-09-10 Nutramax Lab Inc Glusosamine chondroitin and manganese composition for the protection and repair of connective tissue
JPH0710768A (en) 1993-05-24 1995-01-13 Suntory Ltd Hyaluronidase inhibitor containing extract of plant belonging to family leguminosae as active ingredient
CN1096680A (en) 1993-06-19 1994-12-28 金赞星 The medicine of treatment cerebrovascular
JP3529811B2 (en) 1993-06-30 2004-05-24 三省製薬株式会社 External preparation for skin
CN1093914A (en) 1993-07-04 1994-10-26 钟定铨 Ten thousand answer acute throat trouble to loose and manufacture method
JPH0725761A (en) * 1993-07-09 1995-01-27 Kureha Chem Ind Co Ltd Agent for protecting cartilage
GB9317071D0 (en) 1993-08-17 1993-09-29 Univ Strathclyde Flavonoids
CN1101856A (en) * 1993-10-20 1995-04-26 宋海 Chinese medicine spray for curing burn and scald
JP3811198B2 (en) 1993-11-17 2006-08-16 株式会社創研 Antiallergic agent from rice
JP3222672B2 (en) 1993-12-16 2001-10-29 三井農林株式会社 Protein synthesis inhibitor
CN1043406C (en) 1993-12-27 1999-05-19 尚庆英 Method for preparation of compound Qingdaisan powder
JPH07223941A (en) * 1994-02-14 1995-08-22 Nippon Ham Kk Anticomplementary substance
JP3412228B2 (en) 1994-02-23 2003-06-03 松下電器産業株式会社 Gas shut-off device
JPH07242555A (en) 1994-03-08 1995-09-19 Suntory Ltd Glucosyl transferase-inhibiting agent and dental caries-resistant agent and dental caries-resistant food containing the same as active ingredient
JP2711630B2 (en) 1994-04-07 1998-02-10 株式会社大阪薬品研究所 External preparation for skin
WO1996000561A1 (en) * 1994-06-30 1996-01-11 Kyowa Hakko Kogyo Co., Ltd. Hair growth stimulant
TW235922B (en) 1994-07-06 1994-12-11 Amboo Devise Co Ltd A production process of deodorant
JPH0826969A (en) 1994-07-15 1996-01-30 Osaka Yakuhin Kenkyusho:Kk Cosmetic composition
KR960003725A (en) 1994-07-16 1996-02-23 신국현 Treatment of cataracts with golden flavonoids
JPH08104628A (en) * 1994-10-04 1996-04-23 Sumitomo Pharmaceut Co Ltd Inhibitor of matrix metalloprotease
US5545411A (en) 1994-12-16 1996-08-13 Bristol-Myers Squibb Company Method for lowering viscosity by use of gum acacia
KR960040370A (en) 1995-05-01 1996-12-17 강상모 Alcohol Disorder Prevention and Treatment Composition
US5589160A (en) 1995-05-02 1996-12-31 The Procter & Gamble Company Dentifrice compositions
AU689036B2 (en) 1995-05-10 1998-03-19 Kureha Chemical Industry Co., Ltd. Pharmaceutical composition containing substance inhibiting HSP47 production
JP3488325B2 (en) 1995-09-28 2004-01-19 株式会社大和生物研究所 Sasasa tea and its manufacturing method
GB2306321A (en) * 1995-10-24 1997-05-07 Anne Child The use of (+)-catechin in the treatment or prophylaxis of connective tissue disorders
JPH09227374A (en) 1996-02-28 1997-09-02 Taiyo Kagaku Co Ltd Composition for inhibiting production of stress protein
CN1159019C (en) 1996-04-02 2004-07-28 火星有限公司 Extractable compounds and method for making and using the same
JPH09278662A (en) 1996-04-10 1997-10-28 Nof Corp Antiallergic agent
JPH1025238A (en) 1996-07-10 1998-01-27 Mikimoto Pharmaceut Co Ltd Preparation for external use for skin
US5773014A (en) * 1996-10-07 1998-06-30 Bioetica, Inc. Compositions and methods for inhibiting the formation of unwanted skin pigmentation
JPH10130162A (en) 1996-10-31 1998-05-19 Kanebo Ltd Hyaluronic acid decomposition inhibitor, agent for treatment of hyaluronic acid abnormal decomposition disease and cosmetic
US5817299A (en) 1996-11-01 1998-10-06 E-L Management Corp. Non-chemical sunscreen composition
US6197808B1 (en) * 1996-11-18 2001-03-06 Cancer Instititute (Hospital), Chinese Academy Of Medical Sciences Methods for treating hyperplasia
JP3213557B2 (en) * 1996-11-18 2001-10-02 三井農林株式会社 A remedy for condyloma acuminatum containing tea catechin as an active ingredient
US5968973A (en) 1996-11-18 1999-10-19 Cancer Institute (Hospital), Chinese Academy Of Medical Sciences Method for treating hyperplasia
JPH10167960A (en) 1996-12-12 1998-06-23 Les-Bell:Kk Hepatocyte cancer recurrence inhibitor
JPH10182415A (en) 1996-12-20 1998-07-07 Rashieru Seiyaku Kk Cosmetic composition
US5804168A (en) * 1997-01-29 1998-09-08 Murad; Howard Pharmaceutical compositions and methods for protecting and treating sun damaged skin
CN1189365A (en) 1997-01-31 1998-08-05 雷学军 Medicine for preventing Aids virus, treponema pallidum, neisseria gonorrhoeae and trichomonad
US5962517A (en) 1997-01-31 1999-10-05 Murad; Howard Pharmaceutical compositions and methods for treating acne
IN186803B (en) 1997-02-05 2001-11-10 Panacea Biotec Ltd
GB9704904D0 (en) * 1997-03-10 1997-04-30 Riley Fletcher Foundation The Essential oil composition
US5766614A (en) * 1997-03-20 1998-06-16 Yong; Liu Burn treatment compositions containing herbal mix
US20040101576A1 (en) 1997-03-21 2004-05-27 Eiichiro Yagi Immunopotentiators
JPH10287528A (en) 1997-04-07 1998-10-27 Rashieru Seiyaku Kk Cosmetic composition
JP4231559B2 (en) * 1997-04-23 2009-03-04 オリザ油化株式会社 Lipoxygenase inhibitor
PL187310B1 (en) 1997-04-29 2004-06-30 Antoni Gwardys Natural antioxidative preparation
US6243841B1 (en) * 1997-05-30 2001-06-05 Texas Instruments Incorporated Automated test and evaluation sampling system and method
US5886155A (en) * 1997-06-18 1999-03-23 Bioresources International Inc. Purification of miraculin glycoprotein using tandem hydrophobic interaction chromatography
IN1997DE01715A (en) 1997-06-24 2015-07-31 Council Scient Ind Res
US6093403A (en) * 1997-08-01 2000-07-25 Phytocell Research, Inc. Sugar imbalance and diabetes treating herbal formulation
KR19990015612A (en) * 1997-08-07 1999-03-05 한영복 Hepatitis C Therapeutic Composition Containing Mixed Extracts of Hwangbaekpi and Matari Plants
US5886029A (en) * 1997-09-05 1999-03-23 Dhaliwal; Kirpal S. Method and composition for treatment of diabetes
EP0956674A1 (en) 1997-10-02 1999-11-17 NIKKO Co., Ltd. Apparatus for protecting against radiowave transmission from a mobile communication device
CN1062753C (en) 1997-10-21 2001-03-07 陈新学 Medicine for preventing and treating eye diseases
EP1027045A4 (en) * 1997-10-31 2004-12-08 Arch Dev Corp Methods and compositions for regulation of 5-alpha reductase activity
US6696484B2 (en) * 1997-10-31 2004-02-24 University Of Chicago Office Of Technology And Intellectual Property Method and compositions for regulation of 5-alpha reductase activity
JPH11140497A (en) 1997-10-31 1999-05-25 Amyuure:Kk Catechin-containing soap
CA2309150A1 (en) * 1997-11-04 1999-05-14 Pfizer Products Inc. Indazole bioisostere replacement of catechol in therapeutically active compounds
US6221341B1 (en) * 1997-11-19 2001-04-24 Oraceutical Llc Tooth whitening compositions
US6083921A (en) 1998-01-12 2000-07-04 Xu; Kai Jian Pharmaceutical compositions and method of using same
US5922756A (en) * 1998-02-14 1999-07-13 Chan; Marion Man-Ying Method of inhibiting nitric oxide synthase
CN1066055C (en) 1998-03-17 2001-05-23 浙江农业大学 Production method for extracting flavonoid compound extract or powder from bamboo leaf
US5908628A (en) * 1998-05-01 1999-06-01 Hou; Liping Compositions with analgesic, antipyretic and antiinflammatory properties
EP0956867A1 (en) * 1998-05-12 1999-11-17 Franz-Peter Dr. Liebel Use of flavonoid glycosides, tanning agents and microorganisms for the therapy and prophylaxis of diabetes mellitus
FR2778663B1 (en) 1998-05-15 2001-05-18 Coletica NOVEL ESTERS OF FLAVONOIDS, THEIR USE IN COSMETICS, DERMOPHARMACY, PHARMACY AND AGRI-FOOD
US6043694A (en) 1998-06-24 2000-03-28 Siemens Aktiengesellschaft Lock arrangement for a calibrated DLL in DDR SDRAM applications
JP2000044481A (en) 1998-07-30 2000-02-15 Sunstar Inc Preparation for external use for skin
US6080401A (en) * 1998-11-19 2000-06-27 Reddy; Malireddy S. Herbal and pharmaceutical drugs enhanced with probiotics
JP4507027B2 (en) * 1998-12-04 2010-07-21 明治乳業株式会社 MMP inhibitor
US6194469B1 (en) * 1998-12-11 2001-02-27 Board Of Trustees Operating Michigan State Univeristy Method for inhibiting cyclooxygenase and inflammation using cherry bioflavonoids
US20020098207A1 (en) * 1999-02-08 2002-07-25 Daniel H. Maes Cholesterol sulfate compositions for enhancement of stratum corneum function
US6264926B1 (en) * 1999-02-12 2001-07-24 Council Of Scientific And Industrial Research Formulation useful as a natural herbal tooth powder
US6241972B1 (en) * 1999-02-19 2001-06-05 Block Drug Company, Inc. Oral care formulation for the treatment of sensitivity teeth
KR100317112B1 (en) * 1999-03-18 2001-12-22 한영복 Pharmaceutical composition comprising mixed-extract of phellodendron cortex and anemarrhena rhizoma for alalgesic and anti-inflammation
BR0006024A (en) 1999-04-08 2001-11-06 Metagenics Inc Composition for the treatment of inflammation and pain in mammals
US6333304B1 (en) 1999-04-20 2001-12-25 Teresa K. Bath Therapeutic compositions containing glucosamine, collagen and a bioflavanol for repair and maintenance of connective tissue
AU4550100A (en) 1999-05-05 2000-11-21 Herzog, Leslie J. Food product
JP4082823B2 (en) 1999-05-06 2008-04-30 日本メナード化粧品株式会社 Phototoxicity inhibitor
JP4521894B2 (en) * 1999-05-28 2010-08-11 明治乳業株式会社 Matrix metalloproteinase inhibitor
EP1207862A2 (en) * 1999-06-07 2002-05-29 The University Of Sheffield Arthritis treatment
KR20010017481A (en) 1999-08-11 2001-03-05 신상국 A bervage composition for removal an acne and a method of manufacturing therof.
US6264995B1 (en) 1999-10-19 2001-07-24 Thomas Newmark Herbal composition for reducing inflammation and methods of using same
JP2001187744A (en) 1999-10-19 2001-07-10 Tasuku Okuda Cell activation agent and product using the same
WO2001030341A1 (en) 1999-10-25 2001-05-03 Japan As Represented By Director General Of Agency Of National Cancer Center Cyclooxygenase-2 expression inhibitors
KR100390639B1 (en) 2000-01-12 2003-07-07 김미정 Manufacturing Method for Food Composition Containing Extracts of Bamboo Leaves
US6248341B1 (en) * 2000-01-14 2001-06-19 Color Access, Inc. Method of treating topical angiogenesis-related disorders
JP2001220353A (en) 2000-02-07 2001-08-14 Noevir Co Ltd Phospholipase a2 inhibitor
US6290995B1 (en) * 2000-02-08 2001-09-18 Zhao Xinxian Plant drug for preventing cancer II
KR100364383B1 (en) * 2000-02-10 2002-12-18 김호철 Extract of Scutellariae Radix having neuroprotective effects and pharmaceutical preparations containing the same
EP1127572A3 (en) * 2000-02-25 2003-05-02 Basf Aktiengesellschaft Use of flavones for treating cycloxygenase-2 mediated diseases
US20020086070A1 (en) * 2000-03-11 2002-07-04 Kuhrts Eric Hauser Anti-inflammatory and connective tissue repair formulations
TWI233353B (en) 2000-04-11 2005-06-01 Takara Bio Inc Therapeutic or preventive agents containing polyphenols
US20030170186A1 (en) 2000-04-18 2003-09-11 Bernadette Geers Novel flavone glycoside derivatives for use in cosmetics, pharmaceuticals and nutrition
EP1147764A3 (en) 2000-04-19 2002-03-20 Nof Corporation Cosmetic composition
US6475530B1 (en) * 2000-05-31 2002-11-05 Eric H. Kuhrts Methods and compositions for producing weight loss
JP4719372B2 (en) 2000-06-21 2011-07-06 花王株式会社 PPAR-dependent gene transcription activator
US6319523B1 (en) 2000-06-29 2001-11-20 James H. Zhou Composition and method for inhibiting oral bacteria
DE60130466T2 (en) 2000-07-26 2008-06-12 Hououdou Co. Ltd. ANTIPRURITIC COMPOSITIONS AND WOUND HEALING COMPOSITIONS
DE60119534T2 (en) 2000-07-28 2007-05-10 Immupharm Aps USE OF HYDROXYETHYL RUTOSIDES FOR THE TREATMENT OF DETECTION SYMPTOMS, SYMPTOMS OF ALLERGIC RHINITIS AND RESPIRATORY INFECTIONS
JP2002053484A (en) * 2000-08-07 2002-02-19 Asahi Breweries Ltd Lipase inhibitor, and food and drink containing the same
KR100417791B1 (en) 2000-08-14 2004-02-11 미즈코리아 주식회사 Antibiotic and cosmetic compositions containing herb medicines
CN1285202A (en) 2000-08-26 2001-02-28 田积天 Huoqingzhengyang medicine
KR20020031608A (en) 2000-10-19 2002-05-03 이응세 Scutellariae Radix extracts showing a good anti-microbial effect, preparing methods thereof and pharmaceutical compositions containing the same
AU2002239764B2 (en) 2000-11-03 2007-06-21 Proteotech, Inc. Methods of isolating amyloid-inhibiting compounds and use of compounds isolated from uncaria tomentosa and related plants
FR2816839B1 (en) 2000-11-20 2003-09-26 Schwartz Laboratoires Robert PRODUCT PREPARING AND PROTECTING THE SKIN AGAINST UV RAYS, ACTING AGAINST CELLULAR AGING AND EXECUTING AN IMMUNOSTIMULANT ACTION
WO2002047615A2 (en) 2000-12-13 2002-06-20 University Of Rochester Oral compositions and use thereof
WO2002047707A2 (en) * 2000-12-15 2002-06-20 Pharmacia Corporation Selective cox-2 inhibition from non-edible plant extracts
JP2004529079A (en) * 2000-12-15 2004-09-24 ファルマシア・コーポレーション Selective COX-2 inhibition by plant extracts
US6555573B2 (en) * 2000-12-21 2003-04-29 The Quigley Corporation Method and composition for the topical treatment of diabetic neuropathy
US20020146467A1 (en) 2001-02-08 2002-10-10 Jung Kyu Yong Herbal composition for the prevention and treatment of dementia
US6387416B1 (en) * 2001-04-05 2002-05-14 Thomas Newmark Anti-Inflammatory herbal composition and method of use
US6391346B1 (en) * 2001-04-05 2002-05-21 Thomas Newmark Anti-inflammatory, sleep-promoting herbal composition and method of use
JP4901025B2 (en) 2001-06-22 2012-03-21 株式会社ナリス化粧品 Elastase inhibitor
CA2451844A1 (en) 2001-06-27 2003-01-09 Unigen Pharmaceuticals, Inc. Method for generating, screening and dereplicating natural product libraries for the discovery of therapeutic agents
US6685971B2 (en) * 2001-06-28 2004-02-03 Rongxiang Xu Method and composition for repairing and promoting regeneration of mucosal tissue in the gastrointestinal tract
DE10133203A1 (en) 2001-07-07 2003-01-16 Beiersdorf Ag Topical compositions containing catechins and/or their gallate esters, useful e.g. for treating or preventing dry skin or inflammatory conditions of the skin, e.g. eczema, polymorphic light dermatosis or psoriasis
CN1395383A (en) * 2001-07-10 2003-02-05 颜森辉 Intelligent all-bearing expert management system
DE10139793A1 (en) 2001-08-14 2003-02-27 Beiersdorf Ag Use of wogonin for the production of cosmetic or dermatological preparations for the prophylaxis and treatment of inflammatory skin conditions and / or for skin protection in the case of sensitive, dry skin
US7338971B2 (en) * 2001-08-30 2008-03-04 El-Naggar Mawaheb M Treatment of inflammatory, cancer, and thrombosis disorders
KR100486763B1 (en) 2001-09-07 2005-05-03 학교법인고려중앙학원 Extracts of Phyllostachys edulis leaf having antioxidant activity and the process for preparation thereof
JP2003081746A (en) 2001-09-12 2003-03-19 Pola Chem Ind Inc Extension inhibitor of dendrite of melanocyte and cosmetic containing the same
CA2460203A1 (en) * 2001-09-17 2003-03-27 Phytos, Inc. Standardized extracts of scutellaria lateriflora
US20040185124A1 (en) * 2002-10-24 2004-09-23 Hiromichi Hayashi Health food and antitumor agent
US20040057908A1 (en) * 2001-12-13 2004-03-25 Bowen William H. Oral compositions and use thereof
US20030125264A1 (en) * 2001-12-29 2003-07-03 Kimberly-Clark Worldwide, Inc. Methods For Treating Wounds
JP2003212786A (en) 2002-01-16 2003-07-30 Univ Nihon Skin care medicament with bamboo extract component as active ingredient
JP2003212771A (en) 2002-01-23 2003-07-30 Taiyo Kagaku Co Ltd Anti-neissria bacterium composition
US20030166583A1 (en) * 2002-02-22 2003-09-04 Oliver Yoa-Pu Hu Dermal cytochrome P450 1A inhibitors and enhancers
US20030165588A1 (en) 2002-03-01 2003-09-04 Unigen Pharmaceuticals, Inc. Identification of free-B-ring flavonoids as potent COX-2 inhibitors
US20050096281A1 (en) * 2002-03-01 2005-05-05 Unigen Pharmaceuticals, Inc. Formulation of a mixture of Free-B-Ring flavonoids and flavans for use in the prevention and treatment of cognitive decline and age-related memory impairments
US7972632B2 (en) 2003-02-28 2011-07-05 Unigen Pharmaceuticals, Inc. Identification of Free-B-Ring flavonoids as potent COX-2 inhibitors
US7108868B2 (en) * 2002-03-22 2006-09-19 Unigen Pharmaceuticals, Inc. Isolation of a dual cox-2 and 5-lipoxygenase inhibitor from acacia
CA2480332A1 (en) 2002-04-15 2003-10-23 Tetsuo Santo Drinking tea for treating dermatitis
JP4723239B2 (en) * 2002-04-30 2011-07-13 ユニジェン・インコーポレーテッド Formulation of a mixture of free-B-ring flavonoids and flavans as therapeutic agents
US8034387B2 (en) * 2002-04-30 2011-10-11 Unigen, Inc. Formulation of a mixture of free-B-ring flavonoids and flavans for use in the prevention and treatment of cognitive decline and age-related memory impairments
US8945518B2 (en) * 2002-04-30 2015-02-03 Unigen, Inc. Formulation of dual eicosanoid system and cytokine system inhibitors for use in the prevention and treatment of oral diseases and conditions
CN1511549A (en) 2002-12-27 2004-07-14 张小丽 Composition containing scutellaria root for anti-tumor, anti-inflammation and tumor preventing medicine
JP2004244385A (en) 2003-02-14 2004-09-02 Pigeon Corp Mouth wash for infant having anticarious activity
JP2006519231A (en) 2003-02-26 2006-08-24 ユニゲン・ファーマシューティカルス・インコーポレーテッド Formulations for the prevention and treatment of carbohydrate-induced diseases and conditions
EP2213294A1 (en) * 2003-03-27 2010-08-04 Unigen Inc. Use of compositions comprising tricin
CN1798568B (en) 2003-04-04 2011-05-11 尤尼根制药公司 Formulation of dual cycloxygenase (cox) and lipoxygenase (lox) inhibitors for mammal skin care
US7225870B2 (en) * 2003-05-01 2007-06-05 Weatherford/Lamb, Inc. Hydraulic tools for setting liner top packers and method for cementing liners
CN1266144C (en) * 2003-09-01 2006-07-26 上海凯曼生物科技有限公司 Compound of flavonoid as well as application and dosage form of extract product of the compound
EP1684737A4 (en) * 2003-11-07 2009-11-11 Oaky Natural Co Ltd Pharmaceutical composition containing guaiacol derivatives and syringol derivatives extracted from natural plant vinegar
KR100465113B1 (en) 2004-01-15 2005-01-14 주식회사 유니젠 Composition comprising an extract of Bambusoideae plant or tricin isolated therefrom
KR100522579B1 (en) 2004-04-30 2005-10-19 (주) 한약마을 Pharmaceutical composition comprising the extracts of scutellaria root and schizandra fruit mixture thereof having an effect of restraint stress
KR100720971B1 (en) 2004-06-11 2007-05-22 주식회사 유니젠 Composition having Bamboo or Bamboo extract for androgen agonist
US20060008749A1 (en) * 2004-07-08 2006-01-12 Frank Sobel Method for manufacturing of a mask blank for EUV photolithography and mask blank
KR100545304B1 (en) 2004-09-01 2006-05-08 주식회사 유니젠 Composition comprising uncaria genus plant having uncaria gambir, or scutellaria radix and/or green tea extract mixture for suppressing cyclooxygenase or 5-lipoxygenase
US20060140881A1 (en) * 2004-12-22 2006-06-29 Guofeng Xu Oral care compositions containing flavonoids and flavans
US8895084B2 (en) * 2004-12-23 2014-11-25 Colgate-Palmolive Company Oral care composition containing extract of unoxidized Camellia
RU2411941C2 (en) 2005-03-10 2011-02-20 Юниджен Фармасьютикалз, Инк. Preparation based on mixture of flavonoids with free b-cycle and flavanes, as therapeutic agent
KR100720973B1 (en) * 2005-03-18 2007-05-22 주식회사 유니젠 Composition comprising isoorientin for suppressing histamine
CN1686187A (en) 2005-03-23 2005-10-26 贵阳高新瑞得科技开发有限公司 Infantile lung clearing phlegm transforming effervescent granules and its making method
KR100761248B1 (en) 2006-10-12 2007-10-04 주식회사 유니젠 Composition for treating atopic dermatitis comprising extracts of bamboo and scutellaria
US9136202B2 (en) 2012-04-17 2015-09-15 Qualcomm Incorporated Enhanced package thermal management using external and internal capacitive thermal material
US10447702B2 (en) 2017-03-20 2019-10-15 Screening Room Media, Inc. Digital credential tiers

Also Published As

Publication number Publication date
US20030216481A1 (en) 2003-11-20
AU2003228777C1 (en) 2009-08-13
KR20040025884A (en) 2004-03-26
US7674830B2 (en) 2010-03-09
JP4723239B2 (en) 2011-07-13
US20060177528A1 (en) 2006-08-10
US9370544B2 (en) 2016-06-21
DE60328676D1 (en) 2009-09-17
AU2003228777B2 (en) 2009-02-26
HK1073247A1 (en) 2005-09-30
EP2108370A1 (en) 2009-10-14
US20030232763A1 (en) 2003-12-18
CN1649611A (en) 2005-08-03
NZ535988A (en) 2005-09-30
US7514469B2 (en) 2009-04-07
WO2003092599A3 (en) 2004-03-11
US20170071997A1 (en) 2017-03-16
US20140178509A1 (en) 2014-06-26
EP1503778A4 (en) 2007-01-03
EP1503778A2 (en) 2005-02-09
RU2379031C2 (en) 2010-01-20
CA2484192C (en) 2012-10-02
RU2004135069A (en) 2005-07-10
CN101837003A (en) 2010-09-22
AU2003228777A1 (en) 2003-11-17
BRPI0309689A2 (en) 2019-03-12
JP2005529898A (en) 2005-10-06
HK1146712A1 (en) 2011-07-08
CN101837003B (en) 2013-09-04
CA2703698C (en) 2016-11-22
CA2484192A1 (en) 2003-11-13
US9849152B2 (en) 2017-12-26
KR100621234B1 (en) 2006-09-13
CN100544715C (en) 2009-09-30
JP2011068662A (en) 2011-04-07
ATE438393T1 (en) 2009-08-15
WO2003092599A2 (en) 2003-11-13
DK1503778T3 (en) 2009-11-16
EP1503778B1 (en) 2009-08-05
US9655940B2 (en) 2017-05-23
ES2330097T3 (en) 2009-12-04
BRPI0309689B1 (en) 2021-06-29

Similar Documents

Publication Publication Date Title
US9849152B2 (en) Formulation of a mixture of Free-B-ring flavonoids and flavans as a therapeutic agent
US9622964B2 (en) Formulation of dual cycloxygenase (COX) and lipoxygenase (LOX) inhibitors for mammal skin care
US8568799B2 (en) Isolation of a dual COX-2 and 5-lipoxygenase inhibitor from acacia

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20230501