Title of Invention

"A COMPOSITION OF MATTER COMPRISED OF A MISXTURE OF AT LEAST ONE FREE B RING FLAVONOID AND AT LEAST ONE FLAVAN"

Abstract TITLE: FORMULATION OF A MIXTURE OF FREE-B-RING FLAVONOIDS AND FLAVANS AS A THERAPEUTIC AGENT. The present invention provides a novel composition of matteri 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 simulatneously inhibiting the cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO) enzymes, and reducing cox-2 mRNA production. Finally, the presetn 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.
Full Text "A COMPOSITION OF MATTER COMPRISED OF A MIXTURE OF AT LEAST ONE
FREE B RING FLAVONOID AND AT LEAST ONE FLAVAN"
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 prostaglandins, 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 (NSAJDS). 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 prostaglandins 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-1ß (IL-1ß) 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
NSAEDs. 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 autoimmune 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
etal. (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 COX-2
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 al. (2001)
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 NLA, 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 (NFkB) 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:
wherein
R1, R2, R3, R4, and R5 are independently selected from the group consisting of-H,
-OH, -SH, OR, -SR, -NH2, -NHR, -KR2, -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 radicalscavenger 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. 112:127-1331
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-1
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 (PPAR?) 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 NF?B
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 Yakurigaku 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-1ß, IL-6, tumor necrosis factor-a (TNF-a), and interferon-y (IFN-?) (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. 62:331-334), 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
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
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
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
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 µM. (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 carynocarpum, Syzygium 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 µM to 138 µM (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 µM (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 µM, 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 DUD-1 cells (IC50 = 415.3 jjM) (Mutoh et al. (2000) Jpn. J. Cancer Res. £1:686-
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 PGE2
production (Park et al. (2001) Biochem. Biophys. Res. Commun. 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 µM 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 axe present primarily in dry and
arid 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 58-01C:177; Saleem et
al. (1998) Hamdard Midicus. 41:63-67). The bark and pods of A. arabica Willd. contain
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
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;
Gilani et al. (1999) Phytotherapy Research 11:665-669), 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 A1-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 further 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:fiavans 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-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 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:

wherein
R1, R2, R3, R4, and R5 are independently selected from the group consisting of-H, -
OH, -SH, OR, -SR, -NH2, -NHR, -NR2, -NR.3+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
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
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.
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,
Asteraceae, Bignoniaceae, Combretaceae, Compositae, Euphorbiaceae, Labiatae,
Lanranceae, 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, Gnaphalium, Helichrysum, Centaurea,
Eupatorium, Baccharis, Sapium, Scutellaria, Molsa, Colebrookea, Stachys, Origanum,
Ziziphora, Lindera, Actinodaphne, Acacia, Denis, Glycyrrhiza, Millettia, Pongamia,
Tephrosia, Artocarpus, Ficus, Pityrogramma, 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 (¦) 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 (?) 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 (µg/mL). The IC50 for COX-1 was calculated as 0.18
µg/mL/unit of enzyme while the IC50 for COX-2 was calculated as 0.28 µg/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
(?) and ovine COX-2 (¦). The data is presented as percent inhibition of assays without
inhibitor vs. inhibitor concentration (µg/mL). The IC50 for COX-1 was determined to be
0.44 vg/mL/unit of enzyme while that of COX-2 was determined to be 0.28 µg/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 (¦). The data is presented as
percent inhibition of assays without inhibitor vs. inhibitor concentration (µg/mL). The
IC50 for COX-1 was calculated as 0.24 µg/mL/unit of enzyme while the IC50 for COX-2
was calculated as 0.48 µg/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 (¦). The data is presented as percent inhibition of assays without inhibitor
vs. inhibitor concentration (µg/mL). The IC50 for COX-1 was determined to be 0.11
µg/mL/unit of enzyme while the IC50 for COX-2 was determined as 0.42 µg/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 (?) and ovine COX-2 (¦). The data is presented as percent inhibition of assays
without inhibitor vs. inhibitor concentration (µg/mL). The IC50 for COX-1 was calculated
as 0.17 µg/mL/unit of enzyme while the IC50 for COX-2 was determined to be 0.41
µg/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 (?). The data is presented as percent
inhibition of assays without inhibitor vs. inhibitor concentration (µg/mL). The IC50 for 5-
LO was 1.38 µg/mL/unit of enzyme.
Figure 11 depicts graphically a profile of the inhibition of COX-1 and COX-2 by
the Univestin™ 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. 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 (µg/mL). The IC50 for COX-1 was 0.76 µg/mL/unit of enzyme
while the IC50 for COX-2 was 0.80 µg/inL/unit.
Figure 12 depicts graphically a profile of the inhibition of COX-1 and COX-2 by
the Univestin™ composition produced through combination of Free-B-ring flavonoids and
flavans extracts in a ratio of 50:50 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 vs. inhibitor concentration
(µg/mL). The IC50 for COX-1 was 0.38 µg/mL/unit of enzyme while the IC50 for COX-2
was 0.84 (µg/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 (µg/mL). The IC50 for COX-1 was 0.18 (µ/mL/unit of enzyme
while the IC50 for COX-2 was 0.41 µg/mL/unit.
Figure 14 depicts the effect of increasing concentrations of Univestin™ on the
amount of LPS-induced newly synthesized LTB4 (?) 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 µg/mL Univestin™ in non-induced cells to treatment with 3
µg/mL ibuprofen as described in Example 16.
Figure 16 compares the effect of various concentrations of Univestin™ on cox-1
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 Univestin™.
Figure 17 compares the effect of 3µg/mL Univestin™ on cox-l 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. Univestin™ 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 Univestin™ (80:20) ratio of
standardized extracts of Free-B-ring flavonoids to flavans) on the AA injected ankles of
mice (Univestin™ + 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 Univestin™ at a dosage
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 Univestin™ at a dosage
of 500 mg/day.
Figure 22 illustrates graphically the 95% confidence interval for the pain index
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
WOMAC score at baseline, 30, 60 and 90 days of treatment with Univestin™ 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 Univestin™ at a dosage
of 500 mg/day.
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.
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
Univestin™ 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
Univestin™ 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 Univestin™ 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 Univestin™ 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 Univestin™ 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:
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:
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 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.
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 Univestin™ 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 Univestin™ can be from about 1%
to 99% and the concentration of flavans in Univestin™ can be from 99% to 1%. In a
preferred embodiment of the invention, the concentration of total Free-B-ring flavonoids in
Univestin™ is approximately 75% with a baicalin content of approximately 60% of total
weight of the Univestin™; and the concentration of total flavans in Univestin™ is
approximately 10% with a catechin content of approximately 9%. In this embodiment, the
total active components (Free-B-ring flavonoids plus flavans) in Univestin™ 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
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
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 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
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
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
fiavonoids:flavans in the composition of matter is approximately 85:15. m 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:fiavans 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, Compositae, 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, Ulmus, 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 .4. 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 Univestin™, which is comprised of a proprietary blending of two standardized
extracts, which contain Free-B-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 14
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 fiavans 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 fiavans 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 Univestin™. Gene expression data was obtained for the inhibition of
cox-l 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 Univestin™
inhibited cox-2 mRNA production without effecting cox-l gene expression. In addition,
when compared with other COX-2 inhibitor drugs, Univestin™ was able to decrease LPS-
stimulated increases in cox-l 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 Univestin™ 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 Univestin™, 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 fiavans in the range of 10-99%
(based on HPLC) as well as the product Univestin™ 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
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
flavans in the range of 10-99% (based on HPLC) as well as the product Univestin™ 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
Univestin™ 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, Univestin™ (250 mg/day or 500 mg/day) or Celebrex™ (also known as
celecoxib) (200 mg/day). The Univestin™, 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. Celebrex™ is a trade name for a prescription drug that is a COX-2 selective
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,
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 Univestin™ compared to the placebo group.
Further, Univestin™ exhibited a similar effectiveness on pain relieve, better effectiveness
at decreasing stiffness, and marked improvement of physical function compared to the
prescription drug Celebrex™. The greatest significance can be seen in comparing each
dose of Univestin™ 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 Univestin™ 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 Univestin™ 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 Univestin™ dose
showed significantly higher effectiveness compared to the 250 mg/day dose within 90 days
(p=0.038) of treatment.
Univestin™ 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, Univestin™ 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, Univestin™ 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 Univestin™ 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 Univestin™ 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 Univestin™ 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 Univestin™, 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, Univestin™
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 Univestin™ on pain, stiffness and functional
impairment caused by osteoarthritis, Example 21 shows a measurable effect by
Univestin™ 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
Univestin™ 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 mg/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 Univestin™.
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 Univestin™ 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 Univestin™ may affect blood glucose
levels as we{l as its effect on weight loss and BMI. Measurable differences in blood
glucose levels are detected with 30 days of initiating treatment with Univestin™. At 90
days, both the 250 and 500 mg/day Univestin™ 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 Univestin™, 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. Univestin™ 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, Univestin™ 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
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
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
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;
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,
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
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).
The bioassay directed screening process for the identification of specific COX-2
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 µM Heme, 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%.

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 µg/mL and 7 to 80 µg/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 µg/mL, 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.
Example 3. HTP Fractionation of Active Extracts
Organic extract (400 mg) from active plant was loaded onto a prepacked flash,
column. (2 cm ID x 8.2 cm, 10g 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 µL was taken for the
COX inhibition assay.
Aqueous extract (750 mg) from active plant was dissolved in water (5 mL), filtered
through a 1 µm 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 µm 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 µL) 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
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
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 µm
particle size 32-60 µm, 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 µL 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
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: 5 ppm. (DMS0-d6) 8.088 (2H, m, H-3",5"), 7.577 (3H, m, H-
2",4",6"), 6.932 (1H, s, H-8), 6.613 (1H, s, H-3). MS: [M+l]+ = 271m/e. The compound
was identified as baicalein. The IC50 of baicalein against the COX-2 enzyme was
determined to be 10 µg/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 µg/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 µg/mL. To
each well, 150 µL of 100 mM Tris-HCl, pH 7.5 was added along with 10 µL of 22 µM
Hematin diluted in tris buffer, 10 µL 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 µL of 2 mM HN,N"N"-Tetramethyl-p-phenylenediamine
dihydrochloride (TMPD) and 20 µL 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.
The presence and quantity of Free-B-nng 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-18
column (250 x 4.5 mm, 5 pan) a using 1% phosphoric acid and acetonitrile gradient from
80% to 20% in 22 minutes. The Free-B-ring flavonoids were detected using a UV detector
at 254 run and identified based on retention time by comparison with Free-B-ring
flavonoid standards.
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 (Jim particle size 32-
60 /an, 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 µL 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 farther 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 (C10), 156.41 (C8). 1HNMR: d ppm. (DMSO-
d6) 9.150 (1H, s, OH), 8.911 (1H,s, OH), 8.835 (1H, s, OH), 8.788 (1H, s, OH), 6.706
(1H, 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 (1H, d, J=2 Hz, H6), 4.839 (1H, d, J=4 Hz, OH), 4.461 (1H, d,
J=7.3 Hz, H2), 3.798 (1H, m, H3), 2.625 (1H, m, H4b), 2.490 (1H, m, H4a). MS: [M+1]+
= 291 m/e. This compound was identified as catechin.
Compound 2. 13CNMR: d 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
(Cll), 144.39 (C14), 155.72 (C6), 156.19 (C10), 156.48 (C8). 1HNMR: d 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, H21), 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 COX-2
enzymes are 7 µg/mL and 20 µg/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 fiavans 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.
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 THP-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 µL) was plated out in
96-well tissue culture plates and incubated for 2 hours at 37°C and 5% CO2. The media
was then replaced with new HOSC media containing 1 ng/mL IL-1b and incubated
overnight. The media was removed again and replaced with 190 mL HOSC media. Test
compounds were then added in 10 µL of HOSC media and were incubated for 15 minutes
at 37°C. Arachidonic acid in HOSC media (20 mL, 100 /iM) was added and the mixture
was incubated for 10 minutes on a shaker at room temperature. Supernatant (20 /xL) was
transferred to new plates containing 190 µL/well of 100 µM 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
HBSS (25 mL) and added to 96-well plates in 200 mL volumes at 5 x 105 cells/well. The
test compounds in RPMI1640 (10 µL) were then added and incubated for 15 minutes at
37°C. Arachidonic acid in RPMI (20 µL) was then added and the mixture was incubated
for 10 minutes on a shaker at room temperature. Supernatant (20 fiL) was added to ELISA
buffer (190 µL) containing indomethacin (100 µM). The supernatants were then analyzed
by ELISA, as described below.
COX-2 Whole Blood Assay. Peripheral blood from normal healthy donors was
collected by venipuncture. Whole blood (500 µL) 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 µg/mL and cultured overnight at 37°C. The
blood was centrifuged (12,000 x g) and the plasma was collected. Plasma (100 µL) was
added to methanol (400 µL) 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
anti-coagulants and immediately aliquoted into 500 µL 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 µL) was added to methanol (400 µL) 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 µg/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 µL) was added and incubated for 1 hour at
room temperature while shaking. Peroxidase conjugated secondary antibody was added in
a 50 µL 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 µL) 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 µL). The plates were then read at 450
run using a Wallac 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 IC50
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-1. 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.
Example 11. Inhibition of 5-Lipoxvgenase bv 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-HPETE, 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 5-HPETE. 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 µL of 0.17 units/µL potato 5-LO, 20 µL of 1.1 mM AA, 100 µL of oxygen-
sensing chromagen, and 10 µL of purified flavan inhibitor to final concentrations ranging
from 0 to 500 µg/mL. The IC50 for 5-LO inhibition from catechin was determined to be
1.38 µg/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 watermethanol, (4) 40:60
water.methanol, (5) 20:80 watermethanol, (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 fiavan extract generated from solvent extraction with 80%
methanol/water provided the best concentration of flavan components.
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 watenmethanol, (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.
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 HPLC 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.
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
A novel composition of matter, referred to herein as Univestin™ 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
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
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
Univestin™ (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 Univestin™ formulation (50.4 kg) having a blending ratio of 85:15.
Table 9 provides the quantification of the active Free-B-ring flavonoids and flavans of this
specific batch of Univestin™ (Lot#G1702-COX-2), determined using the methods
provided in Examples 7 and 9.
With reference to Table 9, this specific batch of Univestin™ contains 86% total
active ingredients, including 75.7% Free-B-ring flavonoids and 10.3% flavans. Two
different dosage levels of final product in capsule form were produced from this batch of
Univestin™ (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 Univestin™ 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 IC50 Values of COX Enzyme
Inhibitions from Three Formulations of Univestin™
The three different formulations of Univestin™ 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).
Example 16. Measurements of Dose Response and IC50 Values of LO Enzyme Inhibition
from a Formulation of Univestin™
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-1 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#406110) was used to assess the effect of Univestin™ on
newly synthesized levels of LTB4 present in each cell line as a measure of Univestin™"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 µg/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 µg/mL.
Univestin™ and ibuprofen, another known 5-LO inhibitor, were added to the HT-
29 cells at 3 µg/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™ 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 Univestin™ 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 Univestin™ at 1, 3, 10, 30 and 100 |ig/mL and celecoxib, ibuprofen and
acetaminophen at 3 µg/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 (Invitrogen™ 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 Prism®7700 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.
Univestin™ 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 µg/mL of Univestin™, celecoxib, ibuprofen or acetaminophen, only Univestin™ 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 Univestin™
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 Univestin™ with in vivo Mouse Ear Swelling
Model
In order to test whether Univestin™ 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 uL 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 Univestin™ showed a measurable dose
response that tracked with increasing doses of Univestin™, 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 Univestin™ control. The 50 mg/kg dose of Univestin™ was as
effective as the 50 mg/kg dose of another strong anti-inflammatory, indomethacin.
Example 19. Evaluation Efficacy of Univestin™ with in vivo Mouse Ankle Joint Swelling
Model
Since Univestin™ is designed to target joint pain, a solution of 20 µL 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 Univestin™
suspended in olive oil -12 hours before while another group was not given Univestin™.
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 Univestin™. The results are set forth in Figure 19. With
reference to Figure 19, the mice given Univestin™ 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 Univestin™ 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
Hip
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:
The Univestin™ was prepared as described in Example 14. This specific batch of
Univestin™ (lot#G1702-COX-2) contains 86% total active ingredients, including 75.7%
Free-B-ring flavonoids and 10.3% flavans. Celecoxib, also known as Celebrex™, 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
of oral administration for 90 days of the placebo or active compound (Univestin™ 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
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(1):39-46).
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
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
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 Univestin™ 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 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.
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 Univestin™ 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 Univestin™ at both
dosages in reducing stiffness, since the confidence intervals heavily overlapped those of
the placebo. Finally, Univestin™ 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.
Based on these data, the 250 mg/day dose of Univestin™ gave the greatest amount
of weight loss and change in BMI followed by the 500 mg/day dose of Univestin™ 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 Univestin™, may have allowed them to exercise more on their
own accord. Alternatively, Univestin™ 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 Univestin™ after 30 and 90 days of treatment.
Example 22. Clinical Evaluation of the Efficacy of Free-B-ring Flavonoids 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 Univestin™ at 250 and 500 mg/day.
These data suggest that both the 250 and the 500 mg/day doses of Univestin™ are
significantly lowering blood glucose levels over time. This impact may or may not be
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 Univestin™ is acting directly
to improve glucose metabolism by decreasing glucose tolerance or by utilizing
carbohydrates more effectively.
WE CLAIM:
1. A composition of matter comprised of a mixture of at least one Free-B-ring
flavonoid and at least one flavan.
2. The composition as claimed in 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
fiavonoid:flavan to 1:99 of Free-B-ring flavonoid:flavan.
3. The composition as claimed in claim 2 wherein the ratio of Free-B-ring
flavonoid:flavan in the composition of matter is about 85:15.
4. The composition as claimed in 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 glycoside of a single or a combination of
multiple sugars, wherein said glycoside is linked to the 7-hydroxy chromone by a carbon,
oxygen, nitrogen or sulfur, comprising, 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 comprising,
hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride and carbonate.
5. The composition as claimed in 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 glycoside of a
single or a combination of multiple sugars, wherein said glycoside is linked to the 7-
hydroxy chromone by a carbon, oxygen, nitrogen or sulfur comprising, 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 comprising,
but not limited to hydroxyl, chloride, iodide, sulfate, phosphate, acetate, fluoride,
carbonate.
6. The composition as claimed in 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 as claimed in 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 as claimed in 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, Leguminosae, Moraceae, Pinaceae, Pteridaceae, Sinopteridaceae,
Ulmaceae and Zingiberacea.
9. The composition as claimed in 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,
Eupalorium, 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 as claimed in claim 6 wherein said flavan is 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 as claimed in 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.
12. A pharmaceutical composition for alleviating joint pain and stiffness and
improving mobility and physical function comprising an effective amount of a mixture
of Free-B-ring flavonoids and flavan.
13. The composition as claimed in claim 12 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
14. The composition as claimed in claim 12 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramuscular, intraperitoneal and intravenous administration.
15. A pharmaceutical composition for and preventing and treating pathological
conditions related to osteoarthritis and rheumatoid arthritis, comprising an effective
amount of a mixture of Free-B-ring flavonoids and flavans together with a
pharmaceutically acceptable carrier.
16. The composition as claimed in claim 15 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
17. The composition as claimed in claim 15 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
18. A pharmaceutical composition for inhibiting the enzymatic activity of the
cyclooxygenase COX-2 enzyme comprising an effective amount of a mixture of Free-B-
ring flavonoids and flavans.
19. The composition as claimed in claim 18 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
20. The composition as claimed in claim 18 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
21. A pharmaceutical composition for inhibiting the enzymatic activity of the 5-
lipoxygenase (5-LO) enzyme comprising an effective amount of a mixture of Free-B-ring
flavonoids and flavans.
22. The composition as claimed in claim 21 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
23. The composition as claimed in claim 15 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
24. A pharmaceutical composition for simultaneously inhibiting the enzymatic
activity of the COX-2 enzyme and the 5-LO enzyme comprising an effective amount of
a mixture of Free-B-ring flavonoids and flavans.
25. The composition as claimed in claim 24 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
26. The composition as claimed in claim 24 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
27. A pharmaceutical composition for the inhibition of cox-2 mRNA production
comprising an effective amount of a mixture of Free-B-ring flavonoids and flavans.
28. The composition as claimed in claim 27 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
29. The composition as claimed in claim 27 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
30. A pharmaceutical composition for preventing and treating diseases and conditions
mediated by COX-2 and 5-LO pathways comprising an effective amount of a mixture
of Free-B-ring flavonoids and flavans.
31. The composition as claimed in claim 30 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
32. The composition as claimed in claim 30 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
33. The composition as claimed in claim 30 wherein the COX-2 and 5-LO pathways
mediated physiological and pathological conditions are selected from the group
consisting of 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 bowel disease; topical infections caused by virus,
bacteria, fungus, sunburn, thermal burns, contact dermatitis, melanoma and carcinoma.
34. A pharmaceutical composition for reducing blood glucose concentrations
comprising an effective amount of a mixture of Free-B-ring flavonoids and flavans.
35. The composition as claimed in claim 34 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
36. The composition as claimed in claim 34 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
37. A pharmaceutical composition for decreasing body mass index and causing
weight loss comprising an effective amount of a mixture of Free-B-ring flavonoids and
flavans.
38. The composition as claimed in claim 37 wherein the composition is administered
in a dosage selected from 0.01 to 200 mg/kg of body weight.
39. The composition as claimed in claim 37 wherein the routes of the administration
are selected from the group consisting of oral, topical, suppository, intravenous, and
intradermic, intragaster, intramusclar, intraperitoneal and intravenous administration.
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 cyclooxyge-nase-2
(COX-2) and 5-lipoxygenase (S-LO) enzymes, and reducing cax-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.

Documents:

1614-kolnp-2004-granted-abstract.pdf

1614-kolnp-2004-granted-assignment.pdf

1614-kolnp-2004-granted-claims.pdf

1614-kolnp-2004-granted-correspondence.pdf

1614-kolnp-2004-granted-description (complete).pdf

1614-kolnp-2004-granted-drawings.pdf

1614-kolnp-2004-granted-examination report.pdf

1614-kolnp-2004-granted-form 1.pdf

1614-kolnp-2004-granted-form 18.pdf

1614-kolnp-2004-granted-form 3.pdf

1614-kolnp-2004-granted-form 5.pdf

1614-kolnp-2004-granted-gpa.pdf

1614-kolnp-2004-granted-letter patent.pdf

1614-kolnp-2004-granted-reply to examination report.pdf

1614-kolnp-2004-granted-specification.pdf


Patent Number 215533
Indian Patent Application Number 01614/KOLNP/2004
PG Journal Number 09/2008
Publication Date 29-Feb-2008
Grant Date 27-Feb-2008
Date of Filing 29-Oct-2004
Name of Patentee UNIGEN PHARMACEUTICALS INC.
Applicant Address 2660 WILLAMETTE DR, N.E. LACEY, USA.
Inventors:
# Inventor's Name Inventor's Address
1 JIA QI 477 JASPER WAY SUPERIOR COLORADO 80027 USA.
PCT International Classification Number A61K
PCT International Application Number PCT/US03/13463
PCT International Filing date 2003-04-30
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/377, 168 2002-04-30 U.S.A.