CA2503196C - Compositions that treat or inhibit pathological conditions associated with inflammatory response - Google Patents

Compositions that treat or inhibit pathological conditions associated with inflammatory response Download PDF

Info

Publication number
CA2503196C
CA2503196C CA2503196A CA2503196A CA2503196C CA 2503196 C CA2503196 C CA 2503196C CA 2503196 A CA2503196 A CA 2503196A CA 2503196 A CA2503196 A CA 2503196A CA 2503196 C CA2503196 C CA 2503196C
Authority
CA
Canada
Prior art keywords
hops
acid
acids
derived
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA2503196A
Other languages
French (fr)
Other versions
CA2503196A1 (en
Inventor
Matthew L. Tripp
John G. Babish
Jeffrey S. Bland
Gary Darland
Robert Lerman
Daniel O. Lukaczer
Deann J. Liska
Terrence Howell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MetaProteomics LLC
Original Assignee
MetaProteomics LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/464,834 external-priority patent/US20040115290A1/en
Priority claimed from US10/464,410 external-priority patent/US8142819B2/en
Application filed by MetaProteomics LLC filed Critical MetaProteomics LLC
Publication of CA2503196A1 publication Critical patent/CA2503196A1/en
Application granted granted Critical
Publication of CA2503196C publication Critical patent/CA2503196C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/53Lamiaceae or Labiatae (Mint family), e.g. thyme, rosemary or lavender
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Abstract

A natural formulation of compounds that would to modulate inflammation is disclosed. The formulation would also inhibit expression of COX-2, inhibit synthesis of prostaglandins selectively in target cells, and inhibit inflammatory response selectively in target cells. The compositions containing at least one fraction isolated or derived from hops. Other embodiments relate to combinations of components, including at least one fraction isolated or derived from hops, tryptanthrin and conjugates thereof, rosemary, an extract or compound derived from rosemary, a triterpene species, or a diterpene lactone or derivatives or conjugates thereof.

Description

COMPOSITIONS THAT TREAT OR INHIBIT PATHOLOGICAL
CONDITIONS ASSOCIATED WITH INFLAMMATORY RESPONSE
Background of the Invention Field of the Invention The present invention relates generally to compositions that can be used to treat or inhibit pathological conditions associated with tissue-specific activation of inflammation and/or NFxB, to methods of modulating inflammation, including in cells, and to methods of modulating NFxB in cells. More specifically, the invention relates to a composition comprising hops extracts or derivatives thereof or a fraction isolated or derived from hops, which can optionally be combined with a second component, such as rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone species, and tryptanthrin. The invention further relates to methods of using the compositions to inhibit expression of cyclooxygenase-2 (COX-2), inhibit synthesis of prostaglandins selectively in target cells, inhibit inflammatory responses selectively in target cells, and/or inhibit NFxB
activation selectively in target cells.

Description of the Related Art Cyclooxygenase (prostaglandin endoperoxide synthase, EC 1.14.991, COX) catalyzes the rate-limiting step in the metabolism of arachidonic acid to prostaglandin H2 (PGH2), which is further metabolized to various prostaglandins, prostacyclin and thromboxane A2 (c.f. Figure 1). In the early 1990s, it was established that COX exists in two isoforms, commonly referred to as COX-1 and COX-2. It was subsequently determined that the COX-1 and COX-2 proteins are derived from distinct genes that diverged well before birds and mammals. Prostaglandins (PGs) generated via the COX-1 and COX-2 pathways are identical molecules and therefore have identical biological effects. COX-1 and COX-2, however, may generate a unique pattern and variable amounts of eicosanoids; therefore, relative differences in the activation of these isozymes may result in quite dissimilar biological responses.
Differences in the tissue distribution and regulation of COX-1 and COX-2 are now considered crucial for the beneficial as well as adverse effects of COX inhibitors.

The generally held concept (COX dogma) is that COX-1 is expressed constitutively in most tissues whereas COX-2 is the inducible enzyme triggered by pro-inflammatory stimuli including mitogens, cytokines and bacterial lipopolysaccharide (LPS) in cells in vitro and in inflamed sites in vivo.
Based primarily on such differences in expression, COX-1 has been characterized as a housekeeping enzyme and is thought to be involved in maintaining physiological functions such as cytoprotection of the gastric mucosa, regulation of renal blood flow, and control of platelet aggregation. COX-2 is considered to mainly mediate inflammation, although constitutive expression is found in brain, kidney and the gastrointestinal tract. Therefore, it would be desirable to down-regulate tissue-specific or cell-specific expression of COX-2.

Arachidonic acid serves as the primary substrate for the biosynthesis of all PGs.
PGs are ubiquitous hormones that function as both paracrine and autocrine mediators to affect a myriad of physiological changes in the immediate cellular environment.
The varied physiological effects of PGs include inflammatory reactions such as rheumatoid arthritis and osteoarthritis, blood pressure control, platelet aggregation, induction of labor and aggravation of pain and fever. The discovery 30 years ago that aspirin and other non-steroidal analgesics inhibited PG production identified PG
synthesis as a target for drug development. There are at least 16 different PGs in nine different chemical classes, designated PGA to PGI. PGs are part of a larger family of 20-carbon-containing compounds called eicosanoids; they include prostacyclins, thromboxanes, and leukotrienes. The array of PGs produced varies depending on the downstream enzymatic machinery present in a particular cell type. For example, endothelial cells produce primarily PGI2, whereas platelets mainly produce TXA2.

Prostaglandins (PG) are believed to play an important role in maintenance of human gastric mucosal homeostasis. Current dogma is that COX-1 is responsible for PG synthesis in normal gastric mucosa in order to maintain mucosal homeostasis and that COX-2 is expressed by normal gastric mucosa at low levels, with induction of expression during ulcer healing, following endotoxin exposure or cytokine stimulation. It now appears that both COX-1 and COX-2 have important physiological roles in the normal gastric mucosa.

Compounds that inhibit the production of PGs by COX have become important drugs in the control of pain and inflammation. Collectively these agents are known as non-steroidal anti-inflammatory drugs (NSAIDs) with their main indications being osteoarthritis and rheumatoid arthritis. However, the use of NSAIDs, and in particular aspirin, has been extended to prophylaxis of cardiovascular disease. Over the last decade, considerable effort has been devoted to developing new molecules that are direct inhibitors of the enzymatic activity of COX-2, with the inference that these compounds would be less irritating to the stomach with chronic use. Therefore, it would be desirable to inhibit inflammation response selectively in target cells.

U.S. patent application 2002/008607OAl of Kuhrts entitled, "ANTI-INFLAMMATORY AND CONNECTIVE TISSUE REPAIR FORMULATIONS"
describes a hops component that has an IC50-WHMA COX-2/COX-1 ratio ranging from about 0.23 to about 3.33. Example 1 of the application describes a composition containing an extract obtained through supercritical carbon dioxide extraction of whole hops (CO2-extract) comprising 42% humulone.

U.S. Patent No. 6,391,346 entitled, "ANTI-INFLAMMATORY, SLEEP-PROMOTING HERBAL COMPOSITION AND METHOD OF USE" describes an orally administered composition capable of reducing inflammation in animals, while promoting sleep for such animals. The composition contains hydroalcoholic extract of hops and supercritical carbon dioxide extract of hops which are used to promote sleep.

An ideal formulation for the treatment of inflammation would inhibit the induction and activity of COX-2 without inhibiting the synthesis of PGE2 in gastric mucosal cells. However, conventional non-steroidal anti-inflammatory drugs lack the specificity of inhibiting COX-2 without affecting gastric PGE2 synthesis and are at risk to cause damages on the gastrointestinal system, when used for extended periods.
Indeed, even the newly developed, anti-inflammatory drugs such as rofecoxib and celexocib produce untoward gastric toxicity in the form of induced spontaneous bleeding and delay of gastric ulcer healing.

Thus, it would be useful to identify a formulation of compounds that would specifically inhibit or prevent the synthesis of prostaglandins by COX-2 with little or no effect on synthesis of PGE2 in the gastric mucosa. Such a formulation, which would be useful for preserving the health of joint tissues, for treating arthritis or other inflammatory conditions, has not previously been discovered. The term "specific or selective COX-2 inhibitor" was coined to embrace compounds or mixtures of compounds that selectively inhibit COX-2 over COX-1. However, while the implication is that such a calculated selectivity will result in lower gastric irritancy, unless the test materials are evaluated in gastric cells, the term "selective inhibitor" does not carry assurance of safety to gastrointestinal cells. Only testing of compound action in target tissues, inflammatory cells and gastric mucosal cells, will identify those agents with low potential for stomach irritation.

The major problem associated with ascertaining COX-2 selectivity (i.e. low gastric irritancy) is that differences in assay methodology can have profound effects on the results obtained. Depicted in Table 1 are the categories of the numerous in vitro assays that have been developed for testing and comparing the relative inhibitory activities of NSAID and natural compounds against COX-1 and COX-2. These test systems can be classified into three groups: (1) systems using animal enzymes, animal cells or cell lines, (2) assays using human cell lines, or human platelets and monocytes, and (3) currently evolving models using human cells that are representative of the target cells for the anti-inflammatory and adverse effects of NSAID and dietary supplements. Generally, models using human cell lines or human platelets and monocytes are the current standard and validated target cell models have not been forthcoming. A human gastric cell line capable of assessing potential for gastric irritancy is a need.

Table 1. Classification of test systems for in vitro assays assessing COX-2 selectivity of anti-inflammatory compounds . `# ;4`Gi ii =F:,'#~'i'a i33iz `<Ei':?.4` :Sii õ`,.
1TEST SXSTEMS>,.
Tit " r $ !:
, a.. 8~.i.>^.,rad '{.
aJ MAN~3197 ~.~A6P~ S 3. )I ^r ~ ~f ~ ' A
n" R<^tiMr?s .k.
Enzymes Enzymes Human Gastric Mucosa Cells Cells Cells Human Chondrocytes Cell lines Cell lines Human Synoviocytes OTHER SYSTEM VARIABLES
1. Source of arachidonic acid - endogenous or exogenous;
2. Various expression systems for gene replication of COX-1 and COX-2;
3. The presence or absence of a COX-2 inducing agent;
4. COX-2 inducing agents are administered at different concentrations and for different periods of time;
5. Duration of incubation with the drug or with arachidonic acid;
6. Variation in the protein concentration in the medium.

'Adapted from Pairet, M. and van Ryn, J. (1998) Experimental models used to investigate the differential inhibition of cyclooxygenase-1 and cyclooxygenase-2 by 5 non-steroidal anti-inflammatory drugs. Inflamm. Res 47, Supplement 2S93-Sl0l.

The enzymes used can be of animal or human origin, they can be native or recombinant, and they can be used either as purified enzymes, in microsomal preparations,' or in whole-cell assays. Other system variables include the source of arachidonic acid. PG synthesis can be measured from endogenously released arachidonic acid or exogenously added arachidonic acid. In the later case, different concentrations are used in different laboratories.

Second, there are various expression systems for gene replication of recombinant COX-1 and COX-2 enzymes. In addition, the cells transfected with the Cox-1 or Cox-2 gene can be of diverse origins, for instance, insect cell lines or COS
cells. Third, the absence or presence of a COX-2 inducing agent can vary. Cells that are stably transfected with the recombinant enzymes express this enzyme constitutively and no inducing agent is used. This is in fundamental contrast with other cells in which COX-2 has to be induced. Induction of COX-2 is commonly performed using bacterial LPS or various cytokines such as interleukin-1B or tumor necrosis factor.
Additionally, these endotoxins and cytokines are administered at various concentrations.

Fourth, the duration of the incubation with the test agent, the COX-2 inducing agent, or with arachidonic acid varies among different laboratories. These differences can influence the quantitative outcome of the study, because the inhibition of is time dependent. Finally, the protein concentration of the medium can vary;
this is an issue for compounds that can bind avidly to plasma proteins.

A useful assay for COX-2 selectivity would have the following characteristics:
(1) whole cells should be used that contain native human enzymes under normal physiological control regarding expression; (2) the cells should also be target cells for the anti-inflammatory and adverse effects of the compounds; (3) COX-2 should be induced, thereby simulating an inflammatory process, rather than being constitutively expressed; and (4) PG synthesis should be measured from arachidonic acid released from endogenous stores rather than from exogenously added arachidonic acid.
Differences in methodology can explain a dramatic difference in the results obtained for COX inhibition. For example, when assayed against the purified enzyme, ursolic acid exhibited an IC50 of 130 M, far outside of possible physiologically obtainable concentrations [Ringbom, T. et al. (1998) Ursolic acid from Plantago major, a selective inhibitor of cyclooxygenase-2 catalyzed prostaglandin biosynthesis. JNat Prod 61, 1212-1215]. In the RAW 264.7 murine macrophage line, Suh et al. report an IC50 for ursolic acid of approximately [Suh, N., et al. (1998) Novel triterpenoids suppress inducible nitric oxide synthase (iNOS) and inducible cyclooxygenase (COX 2) in mouse macrophages. Cancer Res 58, 717-723]; and in phorbol 12-myristate 13-acetate stimulated human mammary cells, the approximate median inhibitory concentration of ursolic acid was 3.0 M
[Subbaramaiah, K. et al. (2000) Ursolic acid inhibits cyclooxygenase-2 transcription in human mammary epithelial cells. Cancer Res 60, 2399-2404].
No laboratory has, as yet, developed an ideal assay for COX-2 selectivity. The whole cell system most commonly used for Rx and OTC products is the human whole blood assay developed by the William Harvey Institute [Warner, T. D. et al.
(1999) Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis.
Proc Natl AcadSci USA 96, 7563-7568]. To date, this assay format has developed more data supporting clinical relevance than any other. However, new research in the role of constitutive expression of COX-2 in normal gastric mucosa necessitates revisiting the relevance of the use of platelets to model COX-1 inhibition in the absence of COX-2.
The extrapolation of gastrotoxicity from platelet studies is no longer on a sound molecular basis. The validation of a human gastric mucosal cell line for establishing the potential target tissue toxicity of cyclooxygenase inhibitors represents a critical need for the development of safe and effective anti-inflammatory agents.

NF-xB, a heterodimer of the proteins p50 and ReIA, is an inducible eukaryotic DNA binding protein complex that is broadly expressed and plays a pivotal role in regulating multiple biological responses, such as the inflammatory and immune responses in mammalian cells. NF-KB regulate the expression of genes encoding cytokines, chemokines, adhesion molecules, and antimicrobial peptides. Targets of NF--KB include IL-2, the IL-2 receptor, and acute-phase proteins of the liver.
In addition to its role in immune responses, NF-KB activation overrides the apoptotic response to TNF and Fas, allowing for proliferation instead.

As shown in Figure 9, NF--KB is cytoplasmic when inactive, maintained there by IxB. Various stimuli lead to activation of IKK (IiB Kinase), which phosphorylates Ii B, marking it for ubiquitination and degradation. Once IKB is degraded, NF-xB is freed to initiate transcription. Following transcriptional activation of a gene, NF-i .B is also rapidly degraded.

Therefore, it would be useful to identify a composition that would modulate expression or activity of NF-xB at the onset of inflammation to decrease the inflammatory response. Additionally, compositions that act as modulators of NF-KB
can affect a wide variety of disorders in a mammalian body. As a result of inhibiting NFiB, which is a transcription factor for COX-2, the expression of COX-2 can be down-regulated.

An ideal formulation for the treatment of inflammation would inhibit the induction and activity of COX-2 without inhibiting the synthesis of PGE2 in gastric mucosal cells. However, conventional non-steroidal anti-inflammatory drugs lack the specificity of inhibiting COX-2 without affecting gastric PGE2 synthesis and are at risk to cause damages on the gastrointestinal system, when used for extended periods.
Indeed, even the newly developed, anti-inflammatory drugs such as rofecoxib (Vioxx , Merck & Co., Inc.) and celexocib (Celebrex , Pfizer, Inc.) produce untoward gastric toxicity in the form of induced spontaneous bleeding and delay of gastric ulcer healing.

Thus, it would be useful to identify a natural formulation of compounds that would specifically inhibit or prevent the synthesis of prostaglandins by COX-2 with little or no effect on synthesis of PGE2 in the gastric mucosa. Such a formulation, which would be useful for preserving the health of joint tissues, for treating arthritis or other inflammatory conditions, has not previously been discovered. The term "specific or selective COX-2 inhibitor" was coined to embrace compounds or mixtures of compounds that selectively inhibit COX-2 over COX-1. However, while the implication is that such a calculated selectivity will result in lower gastric irritancy, unless the test materials are evaluated in gastric cells, the term "selective COX-2 inhibitor" does not carry assurance of safety to gastrointestinal cells.
Only testing of compound action in target tissues, inflammatory cells and gastric mucosal cells will identify those agents with low potential for stomach irritation.

While glucosamine is generally accepted as being effective and safe for treating osteoarthritis, medical intervention into the treatment of degenerative joint diseases is generally restricted to the alleviation of its acute symptoms. Physicians generally utilize non-steroidal and steroidal anti-inflammatory drugs for treatment of osteoarthritis. These drugs, however, are not suited for long-term therapy because they not only lack the ability to protect cartilage, they can actually lead to degeneration of cartilage or reduction of its synthesis. Moreover, most non-steroidal, anti-inflammatory drugs damage the gastrointestinal system when used for extended periods. Thus, new treatments for arthritis and osteoarthritis combining anti-inflammatory agents with cartilage rebuilding agents are urgently needed.
The joint-protective properties of glucosamine would make it an attractive therapeutic agent for osteoarthritis except for two drawbacks: (1) the rate of response to glucosamine treatment is slower than for treatment with anti-inflammatory drugs, and (2) glucosamine may fail to fulfill the expectation of degenerative remission. In studies comparing glucosamine with non-steroidal anti-inflammatory agents, for example, a double-blinded study comparing 1500 mg glucosamine sulfate per day with 1200 mg ibuprofen, demonstrated that pain scores decreased faster during the first two weeks in the ibuprofen patients than in the glucosamine-treated patients.
However, the reduction in pain scores continued throughout the trial period in patients receiving glucosamine and the difference between the two groups turned significantly in favor of glucosamine by week eight. Lopes Vaz, A., Double-blind clinical evaluation of the relative efficacy of ibuprofen and glucosamine sulphate in the management of osteoarthritis of the knee in outpatients, 8 Cuff. Med Res Opin.

149 (1982). Thus, glucosamine may relieve the pain and inflammation of arthritis, but at a slower rate than the available anti-inflammatory drugs.

An ideal formulation for the normalization of cartilage metabolism or treatment of osteoarthritis would provide adequate chondroprotection with potent anti-inflammatory activity. The optimal dietary supplement for osteoarthritis should enhance the general joint rebuilding qualities offered by glucosamine and attenuate the inflammatory response without introducing any harmful side effects. It should be inexpensively manufactured and comply with all governmental regulations.
However, the currently available glucosamine formulations have not been formulated to optimally attack and alleviate the underlying causes of osteoarthritis and rheumatoid arthritis. Moreover, as with many commercial herbal and dietary supplements, the available formulations do not have a history of usage, nor controlled clinical testing, that might ensure their safety and efficacy.

Therefore, it would be useful to identify a composition that would specifically inhibit or prevent the expression of COX-2 enzymatic activity in inflammatory cells, 5 while having little or no effect on PGE2 synthesis in gastric mucosal cells so that these formulations could be used with no gastrointestinal upset. Furthermore, such formulations should allow for healing of pre-existing ulcerative conditions in the stomach.

Summary of the Invention 10 Thus, it would be useful to identify a formulation of compounds that would modulate an inflammatory response. It would also be useful to identify a formulation of compounds that would modulate NFKB. Such a formulation has widespread applications.

It would also be useful to identify a formulation of compounds that would inhibit expression of COX-2, inhibit prostaglandin synthesis selectively in target cells, or inhibit inflammation response selectively in target cells. For example, it would also be useful to identify a formulation of compounds that would specifically inhibit or prevent the synthesis of prostaglandins by COX-2 in inflammatory cells with little or no effect on PGE2 synthesis in gastric mucosal cells. Such a formulation, which would be useful for preserving the health of joint tissues, for treating arthritis or other inflammatory conditions, has not previously been discovered. Preferably, the formulations have a median effective concentration for COX-2 inhibition in inflammatory cells that is minimally ten times greater than the median effective concentration for the inhibition of PGE2 synthesis in gastric cells. For example, if the median inhibitory concentration for COX-2 of a test formulation was 0.2 gg/mL
in the murine macrophage RAW 264.7, the formulation would not be considered to have low potential for gastric irritancy unless the median inhibitory concentration for PGE2 synthesis in gastric cells was equal to or greater than 2 g/mL.
A preferred embodiment comprises compositions containing at least one fraction isolated or derived from hops (Humulus lupulus). Examples of fractions isolated or derived from hops are alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.
Preferred compounds of fractions isolated or derived from hops, include, but are not limited to, humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. Preferred compounds can also bear substituents, such as halogens, ethers, and esters.
Other embodiments relate to combinations of components. One embodiment relates to compositions that include, as a first component, an active ingredient isolated or derived from an extract of hops and as a second component at least one member selected from the group consisting of rosemary (Rosmarinus officinalis L.), an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof, and tryptanthrin or conjugates thereof. Another embodiment relates to compositions that include, as a first component, tryptanthrin or conjugates thereof and as a second component at least one member selected from the group consisting of an active ingredient isolated or derived from an extract of hops, rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, and a diterpene lactone species or derivatives or conjugates thereof.
As used herein, an extract refers to an extract containing an active ingredient that effects an activity, for example, inhibiting inflammation, inhibiting inducibility or activity of COX-2, inhibiting prostaglandin synthesis, modulating NFKB, and the like.
Preferred compositions can inhibit the inducibility or activity of COX-2.
Compositions of the invention can also function to modulate NFiB. Preferred compositions also can inhibit prostaglandin synthesis selectively in target cells.
Preferred compositions also can inhibit inflammation response selectively in target cells. As used herein, an extract refers to an extract containing an active ingredient that effects an activity, for example, inhibiting inflammation, inhibiting inducibility or activity of COX-2, inhibiting prostaglandin synthesis, modulating NFxB, and the like.

The compositions have widespread applications. Preferred compositions can be useful for treating conditions, such as cancer, autoimmune diseases, inflammatory diseases, or neurological diseases, and obesity. Preferred compositions are also believed to be useful for treating conditions, such as HIV-1 infections, rhinovirus infections, and cardiovascular diseases.

Preferred compositions would be useful for, but not limited to, the treatment of inflammation in a subject, and for treatment of other inflammation-associated disorders, such as an analgesic in the treatment of pain and headaches, or as an antipyretic for the treatment of fever. Preferred compositions would be useful to treat arthritis, including but not limited to rheumatoid arthritis, spondyloathopathies, gouty arthritis, osteoarthritis, systemic lupus erythematosis, and juvenile arthritis.

Preferred compositions would be useful in the treatment of asthma, bronchitis, menstrual cramps, tendonitis, bursitis, and skin-related conditions such as psoriasis, eczema, burns and dermatitis. Preferred compositions also would be useful to treat gastrointestinal conditions such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis and for the, prevention or treatment of cancer such as colorectal cancer.

Further, preferred compositions would be useful in treating inflammation in such diseases as vascular diseases, migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodma, rheumatic fever, type I
diabetes, myasthenia gravis, multiple sclerosis, sacoidosis, nephrotic syndrome, Behchet's syndrome, polymyositis, gingivitis, hypersensitivity, swelling occurring after injury, myocardial ischemia, peridontal disease, fibromyalgia, atopic dermatitis, insulitis and the like.

Additionally, preferred compositions would also be useful in the treatment of ophthalmic diseases, such as retinopathies, conjunctivitis, uveitis, ocular photophobia, and of acute injury to the eye tissue. Preferred compositions would also be useful in the treatment of pulmonary inflammation, such as that associated with viral infections and cystic fibrosis.

Preferred compositions would also be useful for the treatment of certain nervous system disorders such as cortical dementias including Alzheimer's disease. As inhibitors of COX-2 mediated biosynthesis of PGE2 in inflammatory cells, these compositions would also be useful in the treatment of allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous system damage resulting from stroke, ischemia and trauma.

Preferred embodiments further provides a composition to increase the rate at which glucosamine or chondrotin sulfate function to normalize joint movement or reduce the symptoms of osteoarthritis.

Preferred embodiments also provide for methods of identifying compositions that would specifically inhibit or prevent the synthesis of prostaglandins by COX-2 in inflammatory cells with little or no effect on PGE2 synthesis in gastric mucosal cells.
In addition, the compositions of the invention would also be useful for the treatment of obesity, syndrome X, and the like, by reversing hyperglycemia, hyperinsulinemia, and dyslipidermia by normalizing activation of IkkB kinase beta and hence properly modulating its effect on transcription of NFicB-activated genes, effectively breaking the cytokine (for example, tumor necrosis factor alpha (TNFa) feed-back loop leading to these conditions and diseases.

Brief Description of the Drawings Figure 1 depicts the induction of cyclooxygenase-2 and the metabolism of arachidonic acid to prostaglandins and other eicosanoids by the cyclooxygenase enzymes. The action of non-steroidal anti-inflammatory agents is through direct inhibition of the cyclooxygenase enzymes.

Figure 2 shows an outline of fractions and compounds that can be obtained from hops.
Figure 3 illustrates [A] the alpha-acid genus (AA) and representative species humulone (R= -CH2CH(CH3)2), cohumulone (R=, -CH(CH3)2), and adhumulone (R=
-CH(CH3)CH2CH3); [B] the isoalpha acid genus (IAA) and representative species isohumulone (R= -CH2CH(CH3)2), isocohumulone (R=, -CH(CH3)2), and isoadhumulone (R= -CH(CH3)CH2CH3); [C] the reduced isomerized isoalpha acid genus (RIAA) and representative species dihydro-isohumulone (R= -CH2CH(CH3)2) dihydro-isocohumulone (R=, -CH(CH3)2), and dihydro-adhumulone (R= -CH(CH3)CH2CH3); [D] the tetra-hydroisoalpha acid genus (THIAA) and representative species tetra-hydro-isohumulone (R= -CH2CH(CH3)2), tetra-hydro-isocohumulone ((R=, -CH(CH3)2), and tetra-hydro-adhumulone (R= -CH(CH3)CH2CH3); [E] and the hexa-hydroisoalpha acid (HHIAA) genus with representative species hexa-hydro-isohumulone (R= -CH2CH(CH3)2) hexa-hydro-isocohumulone (R=, -CH(CH3)2), and hexa-hydro-adhumulone (R= -CH(CH3)CH2CH3).

Figure 4 illustrates the chemical structure of tryptanthrin.

Figure 5 illustrates the general chemical structures of the triterpene genus [A] and ursolic acid [B] and oleanolic acid [C] as a species within that genus.

Figure 6 [A] shows representative immunoblots demonstrating constitutive COX-1 and COX-2 expression in AGS human gastric mucosal cells. The AGS human gastric cell line was cultured in 6-well plates at 37 C with 5% CO2 in a humidified incubator for 24 hours. Cells were lysed on ice in lysis buffer and protein concentration determined. Fifty gg of cell lysate were solubilized, fractionated on a 10% polyacrylamide gel containing sodium dodecylsulfate (SDS), and transferred onto a nitrocellulose membrane. The membranes were incubated in a blocking buffer and then incubated with the respective primary antibody for 1 h at room temperature.
Following primary antibody incubation, the blots were washed three times with Tris-buffered saline and then incubated with the secondary antibody for 1 h.
Protein bands were visualized using enhanced chemiluminescence. Figure 6 [B] shows densitometric analysis of the immunoblots shown in Figure 6A. Band intensities were evaluated through densitometric analysis, computed using ScanAnalysis software (BIOSOFT, Ferguson, MO), and recorded as arbitrary Density Units (DU).

Figure 7 [A] shows the percent inhibition of PGE2 synthesis in LPS-stimulated RAW 264.7 cells by plasma samples from a human volunteer receiving 880 mg t.i.d.
5 of a test hops derivative formulation. White bars are means of raw data and dark bars are those means computed with the elimination of outliers (never more than two of the eight replicates). The gel capsules of the test formulation contained 200 mg reduced isomerized alpha-acids, 200 mg rosemary extract and 40 mg oleanolic acid.
Figure 7[B] is an estimate of the plasma concentrations of test material at each post-dosing 10 time capable of inhibiting PGE2 synthesis in LPS-stimulated RAW 264.7 cells assuming a constant 5:5:1 ratio of components.

Figure 8 illustrates the induction of PGE2 synthesis by mite allergen in A549 pulmonary cells treated for 24 hours.

Figure 9 shows a pathway of activation of NF--KB. In the cytoplasm, NF-xB is 15 inhibited by IKB. An upstream activating signal may cause phosphorylation of Iid3 by IKK (IKB kinase). This triggers the degradation of 11B through the ubiquitin system.
Once freed from IiB, the free NF-KB can then translocate to the nucleus and activate transcription.

Figure 10 shows dose-related inhibition of PGE2 biosynthesis for RIAA
(shaded bar) and IAA (white bar) treated RAW 264.7 cells following overnight LPS-stimulation prior to the addition of test material.

Figure 11 shows comparison of log IC5o ratios and ranking of potential gastropathy. Log IC5o ratios using the William Harvey Modified Assay (WI MA) are expressed WHMA COX-11WHMA COX-2 from Warner, et al. (white bars) and Mitchell, et al. (dark bars) are shown in [A} with log IC50 ratios (AGS/WHMA
COX-2) for AGS cells treated with A23187 [B], 100 M arachidonic acid [C], or 5 gM
arachidonic acid [D]. Values to the right of 0 indicate decreasing probability of gastropathy, whereas values to the left of 0 indicate increasing probability of gastropathy.

Detailed Description of the Preferred Embodiment The present invention relates to the discovery that that a supragenus of components isolated or derived from hops and other compounds result in tissue-specific or cell-specific inhibition of COX-2 expression. Importantly, these compounds are not believed to directly inhibit COX-2 or other enzymes within the prostaglandin synthesis pathway. Preferred embodiments provide compositions and methods for inhibiting COX-2 expression, inhibiting prostanglandin synthesis selectively in target tissues or cells, or inhibiting inflammation response selectively in target tissues or cells. Compositions and methods of the invention can also modulate NFKB.

A preferred embodiment comprises compositions containing fractions or compounds isolated or derived from hops. Examples of fractions isolated or derived from hops are alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops. Preferred compounds of the fractions isolated or derived from hops can be represented by a supragenus below:
R O O

R"
T HO
R' OH
X
(Supragenus), z wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3i and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and it orbital, with the proviso that if one of R, T, X, or Z is a 7t orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.
Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:
O o R"
HO
R' OH
(Genus A), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:

O O

R"
HO
R' OH
(Genus B), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

Examples of preferred compounds of an ingredient isolated or derived from hops, include, but are not limited to, humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. The preferred compounds can bear substituents, as shown in the formula above.

Another embodiment comprises composition containing tryptanthrin and conjugates thereof.

Other embodiments relate to combinations of components. In particular embodiments, the compositions of the invention can function to specifically inhibit COX-2 expression, to modulate NFiB, to inhibit prostaglandin synthesis selectively in target cells, or to inhibit inflammation response selectively in target cells. The compositions can exhibit synergistic activity.

One embodiment relates to compositions that include, as a first component, an active ingredient isolated or derived from an extract of hops and as a second component at least one member selected from the group consisting of rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof, and tryptanthrin or conjugates thereof. Another embodiment relates to compositions that include, as a first component, tryptanthrin or conjugates thereof and as a second component at least one member selected from the group consisting of an active ingredient isolated or derived from an extract of hops, rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, and a diterpene lactone species or derivatives or conjugates thereof.

As used herein, the term "dietary supplement' 'refers to compositions consumed to affect structural or functional changes in physiology. The term "therapeutic composition" refers to any compounds administered to treat or prevent a disease.

As used herein, the term "effective amount' 'means an amount necessary to achieve a selected result. Such an amount can be readily determined without undue experimentation by a person of ordinary skill in the art.

As used herein, the term "substantial" means being largely but not wholly that which is specified.

As used herein, the term "COX inhibitor" refers to a composition of compounds that is capable of inhibiting the activity or expression of COX-2 enzymes or is capable of inhibiting or reducing the severity, including pain and swelling, of a severe inflammatory response.

As used herein, the terms "derivatives" or a matter "derived" refer to a chemical substance related structurally to another substance and theoretically obtainable from it, i.e. a substance that can be made from another substance. Derivatives can include compounds obtained via a chemical reaction.

As used herein, the term "inflammatory cell" refers to those cellular members of the immune system, for example B and T lymphocytes, neutrophils or macrophages involved in synthesis of prostaglandins in response to inflammatory signals such as interleukins, tumor necrosis factor, bradykinin, histamine or bacterial-derived components.
As used herein, the term "target cells" refers to that cell population in which the inhibition of PGE2 or other prostaglandin synthesis is desired, such as inflammatory cells, tumor cells, or pulmonary cells. Alternatively, "non-target cells"
refers to that cell population in which the inhibition of PGE2 or other prostaglandin synthesis is not desired, such as the gastric mucosal, neural or renal cells.

As used herein, the term "hop extract" refers to the solid material resulting from (1) exposing a hops plant product to a solvent, (2) separating the solvent from the hops plant products, and (3) eliminating the solvent.

As used herein, the term "solvent" refers to a liquid of aqueous or organic nature possessing the necessary characteristics to extract solid material from the hop plant product. Examples of solvents would include, but not limited to, water, steam, superheated water, methanol, ethanol, hexane, chloroform, liquid CO2, liquid N2 or any combinations of such materials.

As used herein, the term "CO2 extract" refers to the solid material resulting from exposing a hops plant product to a liquid or supercritical CO2 preparation followed by removing the CO2.

As used herein, the term "spent hops" refers to the solid and hydrophilic residue from extract of hops.

As used herein, the term "alpha acid" refers to compounds refers to compounds collectively known as humulones and can be isolated from hops plant products including, among others, humulone, cohumulone, adhumulone, hulupone, and isoprehumulone.

As used herein, the term "isoalpha acid" refers to compounds isolated from hops plant products and subsequently have been isomerized. The isomerization of alpha 5 acids can occur thermally, such as boiling. Examples of isoalpha acids include, but are not limited to, isohumulone, isocohumulone, and isoadhumulone.

As used herein, the term "reduced isoalpha acid" refers to alpha acids isolated from hops plant product and subsequently have been isomerized and reduced, including cis and trans forms. Examples of reduced isoalpha acids (RIAA) include, 10 but are not limited to, dihydro-isohumulone, dihydro-isocohumulone, and dihydro-adhumulone.

As used herein, the term "tetra-hydroisoalpha acid" refers to a certain class of reduced isoalpha acid. Examples of tetra-hydroisoalpha acid (THIAA) include, but are not limited to, tetra-hydro-isohumulone, tetra-hydro-isocohumulone and tetra-15 hydro-adhumulone.

As used herein, the term "hexa-hydroisoalpha acid" refers to a certain class of reduced isoalpha acid. Examples of hexa-hydroisoalpha acids (HHIAA) include, but are not limited to, hexa-hydro-isohumulone, hexa-hydro-isocohumulone and hexa-hydro-adhumulone.
20 As used herein, the term "beta-acid fraction" refers to compounds collectively known as lupulones including, among others, lupulone, colupulone, adlupulone, tetrahydroisohumulone, and hexahydrocolupulone.

As used herein, the term "essential oil fraction" refers to a complex mixture of components including, among others, myrcene, humulene, beta-caryophyleen, undecane-2-on, and 2-methyl-but-3-en-ol.

As used herein, "conjugates" of compounds means compounds covalently bound or conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, acetate, and glutathione.
Preferably, the mono- or di- saccharide is a member selected from the group consisting of glucose, mannose, ribose, galactose, rhamnose, arabinose, maltose, and fructose.

As used herein, the term "fats" refers to triacylglyerol esters of fatty acids.

As used herein, the term "waxes" refers to triacylglycerol ethers of or esters of extremely long chain (>25 carbons) fatty alcohols or acids.

HOPS
Hop extraction in one form or another goes back over 150 years to the early nineteenth century when extraction in water and ethanol was first attempted.
Even today an ethanol extract is available in Europe, but by far the predominant extracts are organic solvent extracts (hexane) and CO2 extracts (supercritical and liquid).

(typically at 60 bars pressure and 50 to 10 C) is in a liquid state and is a relatively mild, non-polar solvent highly specific for hop soft resins and oils. Beyond the critical point, typically at 300 bars pressure and 60 C, CO2 has the properties of both a gas and a liquid and is a much stronger solvent. The composition of the various extracts is compared in Table 2.

Table 2. Hop Extracts (Percent W/W) Component Hops Organic Solvent Super-Critical CO2 Liquid CO2 Total resins 12 - 20 15 - 60 75 - 90 70 - 95 Alpha-acids 2-12 8-45 27 - 55 30 -60 Beta-acids 2-10 8-20 23 - 33 15 - 45 Essential oils 0.5-1.5 0-5 1-5 2-10 Hard resins 2-4 2-10 5-11 None Tannins 4-10 0.5 - 5 0.1-5 None Waxes 1-5 1-20 4-13 0-10 Water 8-12 1-15 1-7 1-5 At its simplest, hop extraction involves milling, pelleting and re-milling the hops to spread the lupulin, passing a solvent through a packed column to collect the resin components and finally, removal of the solvent to yield a whole or "pure"
resin extract.

The main organic extractants are strong solvents and in addition to virtually all the lupulin components, they extract plant pigments, cuticular waxes, water and water-soluble materials.

Supercritical CO2 is more selective than the organic solvents and extracts less of the tannins and waxes and less water and hence water-soluble components. It does extract some of the plant pigments like chlorophyll but rather less than the organic solvents do. Liquid CO2 is the most selective solvent used commercially for hops and hence produces the most pure whole resin and oil extract. It extracts hardly the hard resins or tannins, much lower levels of plant waxes, no plant pigments and less water and water-soluble materials.

As a consequence of this selectivity and the milder solvent properties, the absolute yield of liquid CO2, extract per unit weight of hops is less than when using the other mentioned solvents. Additionally, the yield of alpha acids with liquid CO2 (89-93%) is lower than that of supercritical CO2 (91-94%) or the organic solvents (93-96%).
Following extraction there is the process of solvent removal, which for organic solvents involves heating to cause volatilization. Despite this, trace amounts of solvent do remain in the extract. The removal of CO2, however, simply involves a release of pressure to volatize the C02-As shown in Figure 2, hops CO2 extracts can be fractionated into components, including hops oils, beta acids, and alpha acids. Hops oils include, but not limited to, humulene, beta-caryophyllene, mycrene, farnescene, gamma-cadinene, alpha-selinene, and alpha-cadinene. Beta acids include, but are not limited to, lupulone, colupulone, adlupulone, tetrahydroisohumulone, and hexahydrocolupulone, collectively known as lupulones. Beta acids can be isomerized and reduced. Beta acids are reduced to give tetra-beta acids. Alpha acids include, but are not limited to, humulone, cohumulone, adhumulone, hulupone, and isoprehumulone. Alpha acids can be isomerized to give isoalpha acids. Iso-alpha acids can be reduced to give reduced-isoalpha acids, tetra-hydroisoalpha acids, and hexa-hydroisoalpha acids.

A preferred embodiment comprises compositions containing fractions or compounds isolated or derived from hops. Examples of fractions isolated or derived from hops are alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops. Preferred compounds of the fractions isolated or derived from hops can be represented by a supragenus below:

R"
T HO
R' OH
X
(Supragenus), Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and orbital, with the proviso that if one of R, T, X, or Z is a a orbital, then the adjacent R, T, X, or Z is also a 7t orbital, thereby forming a double bond.
Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:

o O

R"
HO
R' OH
(Genus A), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.
Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:

O O

R"
HO

(Genus B), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

As shown in Figure 3, examples of preferred compounds of an ingredient isolated or derived from hops, include, but are not limited to, humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. The preferred compounds can bear substituents, as shown in the formula above.

The identification of humulone from hops extract as an inhibitor of bone resorption is reported in Tobe, H. et al. 1997. (Bone resorption Inhibitors from hop extract. Biosci. Biotech. Biochem 61(1)158-159.) Tobe et al. merely discloses the use of humulone, cohumulone, adhumulone, isohumulone, isocohumulone, and isoadhumulone for treating osteoporosis. Later studies by the same group characterized the mechanism of action of humulone as inhibition of COX-2 gene transcription following TNFalpha stimulation of MC3T3, El cells [Yamamoto, K.
2000. Suppression of cyclooxygenase-2 gene transcription by humulon of beer hop extract studied with reference to the glucocorticoid receptor. FEBS Letters 465:103-106]. The authors concluded that the action of humulone (also humulon) was similar to that of glucocorticoids, but that humulone did not function through the glucocorticoid receptor. While these results establish that humulone inhibits synthesis in MC3T3 cells (osteoblasts) at the gene level, one skilled in the art would not assume that these results would necessarily occur in immune inflammatory cells or other cell lines. Example 5 herein demonstrates the high degree of tissue selectivity of hops compounds and derivatives.

Preferred embodiments provide compositions and methods for inhibiting 5 expression of COX-2, modulating NFKB tissue specifically and cell specifically, inhibiting synthesis of prostaglandins selectively in target cells, and inhibiting inflammatory response selectively in target cells. Preferred methods comprise a step of administering to a mammal a composition of the preferred embodiments.
Preferred embodiments comprise a fraction isolated or derived from hops. A certain 10 composition comprises alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, or spent hops from hops extract or derivatives thereof. Preferred compounds of the fractions isolated or derived from hops can be represented by a supragenus below:

R"
T HO
R' OH
X
(Supragenus), Z
15 wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I and 7t orbital, with the proviso that if one of R, T, X, or Z is a a orbital, then the adjacent R, T, X, or Z is 20 also a t orbital, thereby forming a double bond. Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:

HO
R' OH
(Genus A), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3. Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:
O o R"
HO
R' OH
(Genus B), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3. The preferred embodiments contemplate compositions comprising beta acids or isomerized or reduced beta acids.
Preferably, the alpha acid, isoalpha acid, reduced isoalpha acid, tetra-hydroisoalpha acid, hexa-hydroisoalpha acid, beta acid, or spent hops of the preferred embodiments is made from hops extract. More preferably, the alpha acid, isoalpha acid, reduced isoalpha acid, tetra-hydroisoalpha acid, hexa-hydroisoalpha acid, beta acid, or spent hops of the preferred embodiments is made from CO2 extract of hops.

Tryptanthrin Preferred embodiments can provide compositions and methods for inhibiting expression of COX-2, modulating NFiB tissue specifically and cell specifically inhibiting synthesis of prostaglandins selectively in target cells, and inhibiting inflammatory response selectively in target cells. Preferred methods comprise a step of administering to a mammal a composition of the preferred embodiments. A
certain composition comprises tryptanthrin and conjugates thereof Depicted in Figure 4, tryptanthrin is a natural compound found in certain herbs, such as Polygonum tinctoriuna and Isatis tinctoria. In traditional Chinese medicine this herb is known as Da Qing Ye or Qing Dai. The herb has demonstrated antibacterial and antiviral activity. It has antipyretic, anti-inflammatory and choleretic properties. Increased phagocytic activity of leukocytes and relaxation of intestinal smooth muscle are additional properties of Qing Dai.

Rosemary Certain of preferred embodiments also include delivering an effective amount of rosemary, rosemary extract, or compounds derived from rosemary with the fraction isolated or derived from hops or tryptanthrin. Preferred additions include, but are not limited to, rosemary, rosemary extract, or those compounds known to be found in rosemary or extracts of rosemary. These include 1,8-cineole, 19-alpha-hydroxyursolic acid, 2-13-hydroxyoleanolic acid, 3-0-acetyloleanolic acid, 3-0-acetylursolic acid, 6-methoxy-luteolin-7-glucoside, 6-methoxyluteolin, 6-methoxyluteolin-7-glucoside, methoxyluteolin-7-methylether, 7-ethoxy-rosmanol, 7-methoxy-rosmanol, alpha-amyrin, alpha-humulene, alpha-hydroxyhydrocaffeic acid, alpha-pinene, alpha-terpinene, alpha-terpinenyl acetate, alpha-terpineol, alpha-thujone, apigenin, apigenin-7-glucoside, curcumene, benzyl-alcohol, l3-amyrenone, l3-amyrin,13-elemene, 13-pinene, betulin**, betulinic acid**, borneol, bornyl-acetate, caffeic acid, camphene, camphor, carnosic acid**, carnosol**, carvacrol**, carvone, caryophyllene, caryophyllene-oxide, chlorogenic acid**, diosmetin**, gamma-terpinene, hesperidin, isoborneol, limonene*, luteolin*, luteolin-3'-O-(3"-O-acetyl)-13-D-glucuronide, luteolin-3'-O-(4"-O-acetyl)-13-D-glucuronide, luteolin-3'-O-13-D-glucuronide, luteolin-7-glucoside, methyl-eugenol, myrcene, neo-chlorogenic acid, nepetin, octanoic acid, oleanolic acid, p-cymene, piperitenone, rosmanol, rosmaric acid, rosmaricine, rosmaridiphenol, rosemarinic acid, rosmarinol, rosmariquinone, sabinene, sabinyl acetate, salicylates, salicylic acid-2-13-D-glucoside, squalene, terpinen-4-ol, terpinolene, thymol, trans-anethole, trans-carveol, ursolic acid, verbenone, and zingiberene. Of the species listed, those containing at least one asterisk (*) are preferred and those containing two asterisks (**) are particularly preferred.
Triterpenes and Diterpene Lactones Certain of preferred embodiments also include delivering an effective amount of a triterpene species or diterpene lactone species with the fraction isolated or derived from hops or tryptanthrin. Preferred triterpenes include oleanolic acid, and ursolic acid. Both ursolic and oleanolic acid are found in a wide variety of botanicals.
Diterpene lactones, such as andrographolide, can be obtained from Andrographis paniculata.

Diterpene lactone species, such as andrographolide, and triterpenes, such as ursolic acid and oleanolic acid, are commonly found in plants and are used for their anti-inflammatory properties. The anti-inflammatory effects of these compounds have been described in the literature since 1960. Their mechanism of action is believed to be due (i) to the inhibition of histamine release from mast cells or (ii) to the inhibition of lipoxygenase and cyclooxygenase activity thereby reducing the synthesis of inflammatory factors produced during the arachidonic acid cascade. Since andrographolide and oleanolic acid have been found to promote the healing of stomach ulcers, it is unlikely that the cyclooxygenase activity that is inhibited is COX-1. Also, andrographolide and oleanolic acid are potent antioxidants, capable of inhibiting the generation of reactive oxygen intermediates and restoring tissue glutathione levels following stress.

For example, botanical sources for ursolic acid can be selected from the group consisting of Adina piluifera, Agrimonia eupatoria, Arbutus unedo, Arctostaphylos uva-ursi, Artocarpus heterophyllus, Catalpa bignoniodes, Catharanthus roseus, Chimaphila umbellata, Cornus florida, Cornus officinalis, Crataegus cuneata, Crataegus laevigata, Crataegus pinnatifida, Cryptostegia grandifolia, Elaeagnus pungens, Eriobotryajaponica, Eucalyptus citriodora, Forsythia suspensa, Gaultheria fragrantissima, Glechoma hederacea, Hedyotis diffusa, Helichrysum angustifolium, Humulus lupulus, Hyssopus officinalis, Ilex paraguariensis, Lavandula angustifolia, Lavandula latifolia, Leonurus cardiaca, Ligustrum japonicum, Limonia acidissima, Lycopus europeus, Malus domestica, Marubium vulgare, Melaleuca leucadendra, Melissa officinalis, Mentha spicata, Mentha x rotundifolia, Monarda didyma, Nerium oleander, Ocimum basilicum, Ocimum basilicum, Ocimum basilicum, Ocimum baslicum, Ocimum canum, Origanum majorana, Origanum vulgare, Plantago asiatica, Plantago major, Plectranthus amboinicus, Prunell vulgaris, Prunella vulgaris, Prunus cerasus, Prunus laurocerasus, Prunus persica, Prunus serotina spp serotina, Psidium guajava, Punica granatum, Pyrus communis, Rhododendron dauricum, Rhododendron ferrugineum, Rhododendron ponticum, Rosmarinus officinalis, Rubus fruticosus, Salvia officinalis, Salvia sclarea, Salvia triloba, Sambucus nigra, Sanguisorba officinalis, Satureja hortensis, Satureja montana, Sorbus aucubaria, Syringa vulgaris, Teucrium chamaedrys Teucrium polium, Teucrium spp, Thevetia peruviana, Thymus serpyllum, Thymus vulgaris, Uncaria tomentosa, Vaccinium corymobosum, Vaccinium myrtillus, Vaccinium vitis idaea, Verbena officinalis, Viburnum opulus var. opulus, Viburnum prunifolium, Vinca minor and Zizyphus jujuba.

Similarly, oleanolic acid is found in Achyranthes aspera, Achyranthes bidentiata, Adina piluifera, Ajpocynum cannabinum, Akebia quinata, Allium cepa, Allium sativum, Arctostaphylos uva-ursi, Calendula officinalis, Catharanthus roseus, Centaurium erythraea, Chenopodium album, Citrullus colocynthis, Cnicus benedictus, Cornus officinalis, Crataegus pinnatifida Cyperus rotundus, Daemonorops draco, Diospyros kaki, Elaeagnus pungens, Eleutherococcus senticosus, Eriobotryajaponica, Eugenia caryophyllata, Forsythia suspensa, Glechoma hederacea, Harpagophtum procumbens, Hedera helix,, Hedyotis diffusa, Helianthus annuus, Hemsleys amabilis, Humulus lupulus, Hyssopus officinalis, Ilex rotunda, Lavandula latifolia, Leonurus cardiaca, Ligustrum japonicum, Ligustrum lucidum, Liquidambar orientalis, Liquidambar styraciflua, Loranthus parasiticus, Luffa aegyptiaca, Melaleuca leucadendra, Melissa officinalis, Mentha spicata, Mentha x rotundifolia, Momordica cochinchinensis, Myristica fragrans, Myroxylon balsamum, Nerium oleander, Ocimum suave, Ociumum basilicum, Olea europaea, Origanum majorana, Origanum vulgare, Paederia scandens, Panax ginseng, Panaxjaponicus, Panax quinquefolius, Patrinia scabiosaefolia, Phytolacca americana, Plantago major, Plectranthus amboinicus, Prunella vulgaris, Prunus cerasus, Psidium guajava, Pulsatilla chinenisis, Quisqualis indica, Rosmarinus officinalis, Salvaia officinalis, Salvia sclarea, Salvia triloba, Sambucus nigra, Satureja hortensis, Satureja montana, Swertia chinensis, Swertia diluta, Swertia mileensis, Syzygium aromaticum, Thymus serpyllum, Thymus vulgaris, Trachycarpus fortunei, Uncaria tomentosa, Vaccinium corymbosum, Vaccinium myrtillus, Viburnum prunifolium, Viscum album, Vitis vinifera, or Zizyphus jujuba.

The preferred botanical sources for ursolic acid is a member selected from the group consisting of Ligustrum japonicum, Plantago asiatica, Plantago major, Prunus 5 species, Uncaria tomentosa, Zizyphus jujuba, Cornus officinalis, Eucalyptus citriodora, Forsythia suspensa, Lavandula latifolia, Malus domestica, Nerium oleander, Ocimum baslicum, Punica granatum, Pyrus communis, Rosmarinus officinalis, Salvia triloba, Sorbus aucubaria, Vaccinium myrtillus, Vaccinium vitis-idaea, and Viburnum opulus var. opulus. The most preferred botanical sources for 10 ursolic acid is a member selected from the group consisting of Ligustrum japonicum, Plantago asiatica, Plantago major, Prunus species, Uncaria tomentosa, and Zizyphus jujuba.

The preferred botanical source for oleanolic acid is a member selected from the group consisting of Eleutherococcus senticosus, Ligustrum japonicum, Ligustrum 15 lucidum, Panax ginseng, Panax japonicus, Panax quinquefolius, Plantago major, Prunella vulgaris, Vitis vinifera, Zizyphus jujuba, Achyranthes bidentiata, Allium cepa, Allium sativum, Cornus officinalis, Daemonorops draco, Forsythia suspensa, Prunus cerasus, Quisqualis indica, Rosmarinus officinalis, Salvia triloba, Syzygium aromaticum, Thymus vulgaris, Uncaria tomentosa, Vaccinium corymbosum, and 20 Vaccinium myrtillus. The most preferred botanical source for oleanolic acid is a member selected from the group consisting of Eleutherococcus senticosus, Ligustrum japonicum, Ligustrum lucidum, Panax ginseng, Panaxjaponicus, Panax quinquefolius, Plantago major, Prunella vulgaris Vitis vinifera and Zizyphus jujuba.

Figure 5 illustrates the general chemical structures of the triterpene genus and 25 ursolic acid and oleanolic acid as a species within that genus.
Representative terpenoids within the genus are 18-a-glycyrrhetinic acid**, 18-13-glycyrrhetinic acid* *, 2-a-3-a-dihydrooxyurs-12-3n-28-onic acid*, 3-a-hydroxyursolic acid*, 3-oxo-ursolic acid*, betulin**, betulinic acid**, celastrol*, eburicoic acid, friedelin*, glycyrrhizin, gypsogenin, oleanolic acid**, oleanolic acid-3-acetate, pachymic acid, pinicolic acid, sophoradiol, soyasapogenol A, soyasapogenol B, tripterin**, triptophenolide*, tumulosic acid, ursolic acid**, ursolic acid-3-acetate, uvaol*, and B-sitosterol. Of the species listed, those containing at least one asterisk (*) are preferred and those containing two asterisks (* *) are particularly preferred.

Examples of diterpene lactone species include, but is not limited to, andrographolide, dehydroandrographolide, deoxyandrographolide, neoandrographolide, selenoandrographolide, homoandrographolide, andrographan, amdrographon, andrographosterin, 14-deoxy-l l-oxoandrographolide, 14-deoxy-I1, 12-didehydroandrographolide, andrographiside, and edelin lactone.

Compositions and Synergistic Combinations Preferred compositions can function to specifically inhibit COX-2 expression, to modulate NFiB, to inhibit prostaglandin synthesis selectively in target cells, or to inhibit inflammation response selectively in target cells. Preferred embodiments include compositions containing fractions or compounds isolated or derived from hops or compositions containing tryptanthrin and conjugates thereof.

A preferred embodiment comprises compositions containing fractions or compounds isolated or derived from hops. Examples of fractions isolated or derived from hops are alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops. Preferred compounds of the fractions isolated or derived from hops can be represented by a supragenus below:
R O O

R"
T HO
R' OH
X
(Supragenus), Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I and a orbital, with the proviso that if one of R, T, X, or Z is a 7c orbital, then the adjacent R, T, X, or Z is also a ''c orbital, thereby forming a double bond.

Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:

R"
HO
R' OH
(Genus A), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

Other preferred compounds of the fractions isolated or derived from hops can be represented by a genus below:

I O O
R"
HO
R' OH
(Genus B), wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

Examples of preferred compounds of an ingredient isolated or derived from hops, include, but are not limited to, humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. The preferred compounds can bear substituents, as shown in the formula above.

Another embodiment comprises composition containing tryptanthrin and conjugates thereof.

Other embodiments relate to combinations of components. Preferred compositions can function to specifically inhibit COX-2 expression, to modulate NFiB, to inhibit prostaglandin synthesis selectively in target cells, or to inhibit inflammation response selectively in target cells, including synergistic effects.
One embodiment relates to compositions that include, as a first component, an active ingredient isolated or derived from an extract of hops and as a second component at least one member selected from the group consisting of rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof, and tryptanthrin or conjugates thereof. Another embodiment relates to compositions that include, as a first component, tryptanthrin or conjugates thereof and as a second component at least one member selected from the group consisting of an active ingredient isolated or derived from an extract of hops, rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof.

Dosage The selected dosage level will depend upon activity of the particular composition, the route of administration, the severity of the condition being treated or prevented, and the condition and prior medical history of the patient being treated.
However, it is within the skill of the art to start doses of the composition at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. If desired, the effective daily dose may be divided into multiple doses for purposes of administration, e.g., two to four separate doses per day. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including body weight, general health, diet, time and route of administration, combination with other compositions and the severity of the particular condition being treated or prevented.

Preferred embodiments include delivering an effective amount of hops fractions, hops compounds, or hops derivatives alone or with in combination with other active ingredients. Preferably, a daily dose of preferred compositions would be formulated to deliver about 0.5 to 10,000 mg of alpha acid, isoalpha acid, reduced isoalpha acid, tetra-hydroisoalpha acid, hexa-hydroisoalpha acid, beta acid, or spent hops per day.
More preferably, an effective daily dose of preferred compositions would be formulated to deliver about 50 to 7500 mg, about 100 to 5000 mg, about 200 to mg, or about 500 to 2000 mg of alpha acids, isoalpha acid, reduced isoalpha acid, tetra-hydroisoalpha acid, hexa-hydroisoalpha acid, beta acid, or spent hops per day.
Preferably, the effective daily dose is administered once or twice a day. A
certain embodiment provides a composition comprising about 0.5 to 800 mg of isoalpha acid or reduced isoalpha acid, more preferably about 50 to 400 mg or about 100 to 200 mg of isoalpha acid or reduced isoalpha acid per day. Another certain embodiment provides a composition comprising about 10 to 3000 mg of reduced isoalpha acid, tetra-hydroisoalpha acid, or hexa-hydroisoalpha acid per day, more preferably about 50 to 2000 mg, about 100 to 1500 mg, or about 200 to 1000 mg, of reduced isoalpha acid, tetra-hydroisoalpha acid, or hexa-hydroisoalpha acid per day. Yet another certain embodiment provides a composition comprising about 50 to 7500 mg of spent hops per day, preferably about 100 to 6000 mg, about 200 to 5000 mg, or about 500 to 3000 mg of spent hops per day.

Preferred embodiments include delivering an effective amount of tryptanthrin or conjugates thereof alone or with in combination with other active ingredients.
Preferably, a daily dose of preferred compositions would be formulated to deliver about 0.0005 to 50 mg tryptanthrin/kg body weight per day. More preferably, an effective daily dose of preferred compositions would be formulated to deliver about 0.01 to 10 mg or about 0.1 to 5 mg tryptanthrin/kg body weight per day.
Preferably, a daily dose of preferred compositions would be formulated to deliver about 0.035 to 3500 mg of tryptanthrin per day. More preferably, an effective daily dose of preferred composition would be formulated to deliver about 0.7 to 700 mg, about 1 to 500 mg or about 10 to 100 mg of tryptanthrin per day. Preferably, the effective daily dose is administered once or twice a day.

Preferred embodiments include delivering an effective amount of rosemary or an extract or compound derived from rosemary in combination with other active 5 ingredients. Preferably, a daily dose of preferred compositions would be formulated to deliver about 0.5 to 5000 mg of rosemary, an extract of rosemary, or rosemary-derived compound per day. More preferably, an effective daily dose of preferred composition would be formulated to deliver about 5 to 2000 mg or about 100 to mg of rosemary, an extract of rosemary, or rosemary-derived compound per day.
10 Preferably, the effective daily dose is administered once or twice a day. A
certain embodiment provides a composition comprising about 75 mg of rosemary extract or rosemary-derived compound or derivative, to be administered once or twice a day.

Preferred embodiments include delivering an effective amount of a triterpene or diterpene lactone species or derivatives or conjugates thereof in combination with 15 other active ingredients. Preferably, a daily dose of preferred compositions would be formulated to deliver about 0.0005 to 50 mg triterpene or diterpene lactone/kg body weight per day. More preferably, an effective daily dose of preferred compositions would be formulated to deliver about 0.01 to 10 mg or about 0.1 to 1 mg triterpene or diterpene lactone/kg body weight per day. Preferably, a daily dose of preferred 20 compositions would be formulated to deliver about 0.035 to 3500 mg of triterpene or diterpene lactone species per day. More preferably, an effective daily dose of preferred composition would be formulated to deliver about 0.7 to 700 mg of triterpene or diterpene lactone species per day. Preferably, the effective daily dose is administered once or twice a day.

25 Preferably, an embodiment provides a composition containing an extract of rosemary and a triterpene, such as oleanolic acid, along with an active ingredient, such as a fraction isolated or derived from hops or tryptanthrin or conjugate thereof.
Preferably, an embodiment provides a composition comprising about 0.01 to 500 mg of rosemary extract and about 0.01 to 500 mg of oleanolic acid. Preferably, an embodiment provides a composition capable of producing concentrations in target tissues of 0.1 to 10 g/g tissue of rosemary extract and about 0.1 to 25gg/g tissue of oleanolic acid.

A composition of preferred embodiments for topical application would contain about 0.001 to 10 weight percent, preferably about 0.1 to 1 weight percent of a hops extract component or derivative or tryptanthrin or conjugate thereof.
Preferred embodiments would produce serum concentrations in the ranges of about 0.0001 to 10 M, preferably about 0.01 to 1 gM of a fraction isolated or derived from hops or tryptanthrin or conjugate thereof. The preferred embodiments for topical application can further comprise an additional ingredient selected from rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof, a fraction isolated or derived from hops or tryptanthrin or conjugates thereof, at concentrations of each component of 0.001 to 10 weight percent, preferably 0.1 to 1 weight percent.
Preferred embodiments would produce serum concentrations in the ranges of about 0.001 to 50 M, preferably about 0.1 gM to 5 gM of the additional ingredient.

A certain composition comprises a first component selected from a fraction isolated or derived from hops and a second component comprising an extract or compound derived from rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof, or tryptanthrin or conjugates thereof.
Preferably, the weight ratio of the first component, i.e. a fraction isolated or derived from hops to the second component, i.e. an extract or compound derived from rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, a diterpene lactone species or derivatives or conjugates thereof, or tryptanthrin or conjugates thereof, is within a range of about 100:1 to about 1:100;
preferably about 50:1 to about 1:50; more preferably about 10:1 to about 1:10.

A certain composition comprises a first component of tryptanthrin and conjugates thereof, and a second component comprising hops fraction, hops compound, hops derivative, rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, or a diterpene lactone species or derivatives or conjugates thereof. Preferably, the weight ratio of the first component, i.e. tryptanthrin and conjugates thereof, to the second component, i.e. hops fraction, hops compound, hops derivative, rosemary, an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, or a diterpene lactone species or derivatives or conjugates thereof, is within a range of about 100:1 to about 1:100; preferably about 50:1 to about 1:50; more preferably about 10:1 to about 1:10; even more preferably about 1:1. It is understood that one skilled in art can readily use the amounts described above or appropriate intermediate amounts that are effective for a desired activity.

Applications of Preferred Compositions As stated previously, the generally held concept (COX dogma) is that COX-1 is expressed constitutively in most tissues whereas COX-2 is the inducible enzyme triggered by pro-inflammatory stimuli including mitogens, cytokines and bacterial lipopolysaccharide (LPS) in cells in vitro and in inflamed sites in vivo.
Based primarily on such differences in expression, COX-1 has been characterized as a housekeeping enzyme and is thought to be involved in maintaining physiological functions such as cytoprotection of the gastric mucosa, regulation of renal blood flow, and control of platelet aggregation. COX-2 is considered to mainly mediate inflammation, although constitutive expression is found in brain, kidney and the gastrointestinal tract. Therefore, it would be desirable to down-regulate expression of COX-2 tissue-specifically or cell-specifically. Such down-regulation can be achieved by modulating NFiB. Examples of target cells include, but are not limited to, inflammatory cells, pulmonary cells, microglia and tumor cells. Examples of nontarget cells include, but are not limited to, gastric mucosal, neural, and renal cells.
The compositions have widespread applications. Preferred compositions can be useful for treating conditions, such as cancer, autoimmune diseases, inflammatory diseases, neurological diseases. Preferred compositions are also believed to be useful for treating conditions, such as HIV-1 infections, rhinovirus infections, and cardiovascular diseases.

Preferred embodiments would be useful for, but not limited to a number of inflammatory conditions and can include conditions associated with tissue-specific activation of NFxB. Thus, the invention includes treatment of inflammation in a subject, and treatment of other inflammation-associated disorders, such as, as an analgesic in the treatment of pain and headaches, or as an antipyretic for the treatment of fever. Additional examples of such preferred embodiments would be useful to treat arthritis, including but not limited to rheumatoid arthritis, spondyloathopathies, gouty arthritis, osteoarthritis, systemic lupus erythematosis, and juvenile arthritis. Such preferred embodiments would be useful in the treatment of asthma, bronchitis, menstrual cramps, tendonitis, bursitis, and skin related conditions such as psoriasis, eczema, burns and dermatitis. Preferred embodiments also would be useful to treat gastrointestinal conditions such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis and for the prevention or treatment of cancer such as colorectal cancer. Preferred embodiments would be useful in treating inflammation in such diseases as vascular diseases, migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodma, rheumatic fever, type I diabetes, myasthenia gravis, multiple sclerosis, sacoidosis, nephrotic syndrome, Behchet's syndrome, polymyositis, gingivitis, hypersensitivity, swelling occurring after injury, myocardial ischemia, periodontal disease, insulitis and the like.

Preferred embodiments would also be useful in the treatment of ophthalmic diseases, such as retinopathies, conjunctivitis, uveitis, ocular photophobia, and of acute injury to the eye tissue. Preferred embodiments would also be useful in the treatment of pulmonary inflammation, such as that associated with viral infections and cystic fibrosis. Preferred embodiments would also be useful in the treatment of asthma. Preferred embodiments would also be useful for the treatment of certain nervous system disorders such as cortical demential including Alzheimer's disease.
Preferred embodiments are useful as anti-inflammatory agents, such as for the treatment of arthritis, with the additional benefit of having significantly less harmful side effects. As inhibitors of COX-2 mediated biosynthesis of PGE2, these compositions would also be useful in the treatment of allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous system damage resulting from stroke, ischemia and trauma. The preferred embodiments would also be useful for the treatment of fibromyalgia.

Since COX-2 can also play a role in the regulation of osteoblastic function, preferred embodiments can also be useful for treating and preventing osteoporosis.
Kanematsu et al. (J Bone Miner Res 1997 Nov; 12(11):1789-96.) discloses that interleukin 1 (IL-1) and tumor necrosis factor alpha (TNF-alpha) have been implicated in the pathogenesis of osteoporosis. These proinflammatory cytokines induce both COX-2 and nitric oxide synthase (iNOS) with the release of PGE2 and NO, respectively. They determined the interaction between COX and NOS pathways and their role in the regulation of osteoblastic function in MC3T3-E1 cells.

According to preferred embodiments, the animal may be a member selected from the group consisting of humans, non-human primates, dogs, cats, birds, horses, ruminants or other warm blooded animals. Preferred embodiments are directed primarily to the treatment of human beings. Administration can be by any method available to the skilled artisan, for example, by oral, topical, transdermal, transmucosal, or parenteral routes.

Besides being useful for human treatment, preferred embodiments are also useful for treatment of other animals, including horses, dogs, cats, birds, sheep, pigs, etc. A
certain formulation for the treatment of inflammation would inhibit the induction and activity of COX-2 with little effect on the synthesis of PGE2 in the gastric mucosa.
Historically, the NSAIDs used for treatment of inflammation lacked the specificity of inhibiting COX-2 without affecting PGE2 synthesis in gastric mucosal cells.
Therefore, these drugs irritated and damaged the gastrointestinal system when used for extended periods.

Preferred compositions can also modulate NF-KB. Modulation of NF-KB can include regulating levels of NF-KB to treat or inhibit a pathological condition in a mammal. For example, abnormal levels, such as increased levels, of NF-KB can be associated with diseases and undesirable conditions. NF-KB is implicated in neuronal 5 survival, inflammatory response, and cancer. NF--KB regulates COX-2 gene expression. Therefore, preferred compositions that modulate NF-KB can also affect the expression of COX-2.

Results presented herein indicate that modulation of NF-KB results in modulation of COX-2 expression in target cells only, without any significant direct inhibition of 10 COX-2 or other enzymes with PG pathway. Therefore, for example, preferred compositions offer the advantage of anti-inflammatory effects without the side effects of damaging gastric mucosa, which are present in many existing NSAIDs.
Existing NSAIDs, such as rofecoxib and celxobib, are supposed to inhibit the synthesis of prostanglandins by selectively inhibiting the COX-2 enzyme. However, side effects 15 still occur with these existing NSAIDs because of lack of total selective inhibition of COX-2. Existing NSAIDs can still be promiscuous and affect enzymes other than COX-2 to result in side effects. By modulating NF-KB, compositions of preferred embodiments act at an upstream position and can inhibit the synthesis of COX-2 selectively in target cells. Without COX-2 in target cells, the synthesis of 20 prostaglandins directed to inflammatory reactions can also be inhibited.
Therefore, the inflammatory reaction can be prevented or halted. While COX-2 in target cells is affected, COX-1 and COX-2 in nontarget cells remain unaffected and continues to maintain physiological functions, such as cytoprotection of gastric mucosa, regulation of renal blood flow, and control of platelet aggregation.

25 Since preferred compositions can affect NF-KB, preferred embodiments can also be useful for treating and preventing a variety of disorders including, but not limited to, autoimmune, inflammatory, neurological and cardiovascular diseases, and cancer.

Preferred embodiments can be useful for treating and preventing a pathological condition associated with tissue-specific activation of NF--KB. NF--KB can be found in numerous cell types and is found to be activated by a wide range of inducers.
Upon activation and transport to the nuclei, NF-KB can initiate or regulate early-response gene transcription by binding to motifs found in the promoter or enhancer regions of specific genes.

The NF-KB response occurs in virtually all cell types in combination with a variety of co-activators. However, because NF-KB alone is not capable of activating its genes when bound to DNA, the exact genes activated will vary depending on the cellular context. Co-activators are believed to link enhancer-bound transcription factors, like NF-KB, to components of the basal transcription machinery, which then transcribe the gene to generate the mRNA copy. Therefore, NF-KB can be modulated tissue- or cell-specifically so as to treat and/or inhibit a variety of pathological conditions.

Therefore, compositions that inhibit or activate specific targets in the NF-KB
pathway provide new approaches in the treatment or prevention of a number of serious diseases, including cancer and inflammatory disorders. NF-kB is a transcription factor that is involved in a range of cellular phenomena, including inflammation, antigen presentation, immunity, cytokine production, apoptosis and cancer. For example, as NF-KB affects pulmonary cells, a pathological condition can be manifested as asthma, and/or other pulmonary conditions. NF-KB is also involved in colorectal, mammary, and prostate conditions, such as cancer, as mediated by PGE2. NF-KB is involved in these cancers and other cancers, as mediated by cell-to-cell adhesion. NF-KB is also involved in HIV-1 replication, colds, and flus.
As stated above, NF-KB has been implicated in conditions, such as autoimmune, inflammatory, neurological and cardiovascular diseases, and cancer.

Formulations Preferred compositions can be administered in the form of a dietary supplement or therapeutic composition. The compositions may be administered orally, topically, transdermally, transmucosally, parenterally, etc., in appropriate dosage units, as desired.

Preferred compositions for dietary application may include various additives such as other natural components of intermediary metabolism, vitamins and minerals, as well as inert ingredients such as talc and magnesium stearate that are standard excipients in the manufacture of tablets and capsules. For example, one embodiment comprises active ingredients of preferred compositions in combination with glucosamine or chondrotin sulfate.

As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, sweeteners and the like. These pharmaceutically acceptable carriers may be prepared from a wide range of materials including, but not limited to, diluents, binders and adhesives, lubricants, disintegrants, coloring agents, bulking agents, flavoring agents, sweetening agents and miscellaneous materials such as buffers and absorbents that may be needed in order to prepare a particular therapeutic composition. The use of such media and agents for pharmaceutically active substances is well known in the art.
Except insofar as any conventional media or agent is incompatible with the active ingredients, its use in preferred compositions is contemplated. In one embodiment, talc, and magnesium stearate are included in the formulation. Other ingredients known to affect the manufacture of this composition as a dietary bar or functional food can include flavorings, sugars, amino-sugars, proteins and/or modified starches, as well as fats and oils.

Dietary supplements, lotions or therapeutic compositions of preferred embodiments can be formulated in any manner known by one of skill in the art.
In one embodiment, the composition is formulated into a capsule or tablet using techniques available to one of skill in the art. In capsule or tablet form, the recommended daily dose for an adult human or animal would preferably be contained in one to six capsules or tablets. However, preferred compositions can also be formulated in other convenient forms, such as an injectable solution or suspension, a spray solution or suspension, a lotion, gum, lozenge, food or snack item.
Food, snack, gum or lozenge items can include any ingestible ingredient, including sweeteners, flavorings, oils, starches, proteins, fruits or fruit extracts, vegetables or vegetable extracts, grains, animal fats or proteins. Thus, preferred compositions can be formulated into cereals, snack items such as chips, bars, gumdrops, chewable candies or slowly dissolving lozenges. Preferred embodiments contemplate treatment of all types of inflammation-based diseases, both acute and chronic. Preferred formulations reduce the inflammatory response and thereby promotes healing of, or prevents further damage to, the affected tissue. A pharmaceutically acceptable carrier can also be used in the preferred compositions and formulations.

Assay using AGS Cell Line In order to identify selective COX-2 drugs, it has been common practice to use the Modified Whole Blood/Cell Assay of T.D. Warner et al., Nonsteroid drug selectivities for cyclooxygenase-1 rather than cyclooxygenase-2 are associated with human gastrointestinal toxicity: A -full in vitro analysis, Proc. Natl. Sci. USA
96:7563-7568(1999). When hop fractions are tested according to this procedure, hops extracts do not yield IC50 values in the necessary g/mL range, since they are not direct inhibitors of COX-2. This lack of direct inhibition of COX-2 was demonstrated by Tobe, H. et al. 1997. (Bone resorption Inhibitors from hop extract. Biosci.
Biotech.
Biochem 61(1)158-159) using purified COX-2 enzyme. Similarly, EXAMPLE 4 of this application demonstrates that, when tested according to the Modified Whole Blood/Cell Assay, hops compounds and derivatives produce median inhibitory concentrations greater than 25 jig/mL. Such high median inhibitory concentrations are pharmacologically unsuitable. Therefore, the Modified Whole Blood Assay as described by Warner is an invalid procedure for formulating potentially therapeutically effective combinations containing hops or hops derivatives.

The discovery of COX-2 has made possible the design of drugs that reduce inflammation without removing the protective PGs in the stomach and kidney made by COX-1. One of our approaches is to screen compositions of the preferred embodiments using in vitro animal cells to assess COX-2 and COX-1 inhibitory activity employing PGE2, which has cytoprotective actions and play a role in maintaining the integrity of the gastrointestinal mucosa, as an endpoint.
Secondarily, different cell types are used to confirm results. The screening process would indicate compositions that have specific COX-2 activity and limited COX-1 inhibition.
Compositions of preferred embodiments can be tested in two cell types: 1) human pulmonary cells or other cell line to determine and identify optimal amounts and ratios for compositions comprising more than one component; and 2) human gastric epithelial cells (AGS cell line), a gastrointestinal tract cell line and a model system for assessing toxicity which is typically related to inhibition of COX-1 which is required for wound healing (such as ulcers). Hence, compositions of preferred embodiments that can inhibit COX-2 or COX-2 induction can be screened by selecting compositions that have low or no activity in AGS cells and good activity in human pulmonary cells or other cell line.

In particular embodiments, the invention provides a composition comprising, as a first component, a fraction derived from hops; and as a second component, at least one member selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone species, and tryptanthrin. The fraction derived from hops can be extracted with C02-The fraction derived from hops can also be selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, and spent hops.

In another embodiment, a composition of the invention can contain a fraction derived from hops comprising a compound of a supragenus having the formula:
R O O

T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and it orbital, with the proviso that if one of R, T, X, or Z is an orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

5 In yet another embodiment, the fraction derived from hops can comprise a compound of Genus A having the formula:

O o R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and 10 wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.
In yet another embodiment, the fraction derived from hops can comprise a compound of Genus B having the formula:

O o R"
HO
R' OH

15 wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In a composition of the invention, the fraction derived from hops can comprise a 20 compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

In a composition of the invention, a second component can be a compound derived from rosemary that is selected from the group consisting of 1,8-cineole, 19-alpha-hydroxyursolic acid, 2-B-hydroxyoleanolic acid, 3-0-acetyloleanolic acid, 3-0-acetylursolic acid, 6-methoxy-luteolin-7-glucoside, 6-methoxyluteolin, 6-methoxyluteolin-7-glucoside, methoxyluteolin-7-methylether, 7-ethoxy-rosmanol, methoxy-rosmanol, alpha-amyrin, alpha-humulene, alpha-hydroxyhydrocaffeic acid, alpha-pinene, alpha-terpinene, alpha-terpinenyl acetate, alpha-terpineol, alpha-thujone, apigenin, apigenin-7-glucoside, curcumene, benzyl-alcohol,13-amyrenone,l3-amyrin, l3-elemene,13-pinene, betulin, betulinic acid, borneol, bornyl-acetate, caffeic acid, camphene, camphor, carnosic acid, carnosol, carvacrol, carvone, caryophyllene, caryophyllene-oxide, chlorogenic acid, diosmetin, gamma-terpinene, hesperidin, isoborneol, limonene, luteolin, luteolin-3'-O-(3"-O-acetyl)-13-D-glucuronide, luteolin-3'-0-(4"-0-acetyl)-13-D-glucuronide, luteolin-3'-0-13-D-glucuronide, luteolin-glucoside, methyl-eugenol, myrcene, neo-chlorogenic acid, nepetin, octanoic acid, oleanolic acid, p-cymene, piperitenone, rosmanol, rosmaric acid, rosmaricine, rosmaridiphenol, rosemarinic acid, rosmarinol, rosmariquinone, sabinene, sabinyl acetate, salicylates, salicylic acid-2-13-D-glucoside, squalene, terpinen-4-ol, terpinolene, thymol, trans-anethole, trans-carveol, ursolic acid, verbenone, and zingiberene.

In another embodiment, the second component can be a compound derived from rosemary that is selected from the group consisting of betulin, betulinic acid, carnosic acid, carnosol, carvacrol, chlorogenic acid, diosmetin, limonene, and luteolin. In still a further embodiement, the second component can be a triterpene species that is selected from the group consisting of 18-a-glycyrrhetinic acid, 18-13-glycyrrhetinic acid, 2-a-3-a-dihydrooxyurs-12-3n-28-onic acid, 3-a-hydroxyursolic acid, 3-oxo-ursolic acid, betulin, betulinic acid, celastrol, eburicoic acid, friedelin, glycyrrhizin, gypsogenin, oleanolic acid, oleanolic acid-3-acetate, pachymic acid, pinicolic acid, sophoradiol, soyasapogenol A, soyasapogenol B, tripterin, triptophenolide, tumulosic acid, ursolic acid, ursolic acid-3-acetate, uvaol, and 13-sitosterol.
Furthermore, the second component can be a triterpene species that is selected from the group consisting of 18-a-glycyrrhetinic acid, 18-13-glycyrrhetinic acid, 2-a-3-a-dihydrooxyurs-12-3n-28-onic acid, 3-a-hydroxyursolic acid, 3-oxo-ursolic acid, betulin, betulinic acid, celastrol, friedelin, oleanolic acid, tripterin, triptophenolide, ursolic acid, and uvaol. In addition, the second component can be tryptanthrin, a triterpene species, or a diterpene lactone species that is conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, acetate, and glutathione.

A composition of the invention can also comprise about 0.5 to 10000 mg of the fraction isolated or derived from hops or about 50 to 7500 mg of the fraction isolated or derived from hops. A composition of the invention can additionally comprise about 0.035 to 3500 mg of tryptanthrin, or about 0.7 to 700 mg of tryptanthrin, wherein the second component is tryptanthrin. A composition of the invention can also comprise about 0.5 to 5000 mg of the second component, or about 5 to 2000 mg of the second component, wherein the second component is selected from the group consisting of rosemary, extract derived from rosemary, and a compound derived from rosemary. Additionally, a composition of the invention can comprise about 0.035 to 3500 mg of a triterpene species, or about 0.7 to 700 mg of a triterpene species, wherein the second component is a triterpene species.

In still another embodiment, a composition can comprise about 0.001 to 10 weight percent of the first component, or about 0.1 to 1 weight percent of the first component.
In addition, a composition can comprise about 0.001 to 10 weight percent of the second component, or about 0.1 to 1 weight percent of the second component. In a composition of the invention, the ratio of the first component to the second component can be in the range of about 100:1 to about 1:100, or about 50:1 to about 1:50. Any of the compositions of the invention can further comprise a pharmaceutically acceptable carrier, and such a composition comprising a pharmaceutically acceptable carrier can be used in the methods of the invention.

The invention also provides a composition comprising as a first component, a fraction isolated or derived from hops; and as a second component, at least one member selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, and tryptanthrin. The fraction isolated or derived from hops can be extracted with CO2. The fraction isolated or derived from hops can be selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

The fraction isolated or derived from hops can comprise a compound of a supragenus having the formula:

R O O
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and 7t orbital, with the proviso that if one of R, T, X, or Z is a 7t orbital, then the adjacent R, T, X, or Z is also a t orbital, thereby forming a double bond.
The fraction isolated or derived from hops can also comprise a compound of Genus A having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.
The fraction isolated or derived from hops can additionally comprise a compound of Genus B having the formula:

O o Rt' HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

The fraction isolated or derived from hops can additionally comprise a compound selected from the group consisting of humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

Furthermore, the second component of such a composition can be a compound derived from rosemary that is selected from the group consisting of 1,8-cineole, 19-alpha-hydroxyursolic acid, 2-13-hydroxyoleanolic acid, 3-0-acetyloleanolic acid, 3-0-acetylursolic acid, 6-methoxy-luteolin-7-glucoside, 6-methoxyluteolin, 6-methoxyluteolin-7-glucoside, methoxyluteolin-7-methylether, 7-ethoxy-rosmanol, methoxy-rosmanol, alpha-amyrin, alpha-humulene, alpha-hydroxyhydrocaffeic acid, alpha-pinene, alpha-terpinene, alpha-terpinenyl acetate, alpha-terpineol, alpha-thujone, apigenin, apigenin-7-glucoside, curcumene, benzyl-alcohol, l3-amyrenone,l3-amyrin, l3-elemene,13-pinene, betulin, betulinic acid, borneol, bornyl-acetate, caffeic acid, camphene, camphor, carnosic acid, carnosol, carvacrol, carvone, caryophyllene, caryophyllene-oxide, chlorogenic acid, diosmetin, gamma-terpinene, hesperidin, isoborneol, limonene, luteolin, luteolin-3'-O-(3"-O-acetyl)-13-D-glucuronide, luteolin-3'-O-(4"-O-acetyl)-13-D-glucuronide, luteolin-3'-O-B-D-glucuronide, luteolin-7-5 glucoside, methyl-eugenol, myrcene, neo-chlorogenic acid, nepetin, octanoic acid, oleanolic acid, p-cymene, piperitenone, rosmanol, rosmaric acid, rosmaricine, rosmaridiphenol, rosemarinic acid, rosmarinol, rosmariquinone, sabinene, sabinyl acetate, salicylates, salicylic acid-2-B-D-glucoside, squalene, terpinen-4-ol, terpinolene, thymol, trans-anethole, trans-carveol, ursolic acid, verbenone, and 10 zingiberene. The second component can also be a compound derived from rosemary that is selected from the group consisting of betulin, betulinic acid, carnosic acid, carnosol, carvacrol, chlorogenic acid, diosmetin, limonene, and luteolin.
Additionaly, the second component can be tryptanthrin that is conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, 15 acetate, and glutathione.

In a particular embodiment, the composition can comprise about 0.5 to 10000 mg or about 50 to 7500 mg of the fraction isolated or derived from hops. In addition, the composition can comprise about 0.35 to 3500 mg of tryptanthrin, or about 0.7 to 700 mg of tryptanthrin, wherein the second component is tryptanthrin. Moreover, the 20 composition can comprise about 0.5 to 5000 mg of the second component, or about 5 to 2000 mg of the second component, wherein the second component is selected from the group consisting of rosemary, extract derived from rosemary, and a compound derived from rosemary. In addition, the composition can comprise about 0.001 to 10 weight percent of the first component, or about 0.1 to 1 weight percent of the first 25 component. Also, the composition can comprise about 0.001 to 10 weight percent of the second component, about 0.1 to 1 weight percent of the second component.
In another embodiment, a ratio of the first component to the second component can be in the range of about 100:1 to about 1:100, or in the range of about 50:1 to about 1:50.
The composition can further comprise a pharmaceutically acceptable carrier.

In still another embodiment, the invention provides a method of modulating inflammatory response in cells, the method comprising contacting the cells with a composition of the invention. For example, the method can be carried out using a composition comprising a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone species, and tryptanthrin. The invention also provides a method of treating or inhibiting a pathological condition in a mammal associated with tissue-specific activation of inflammation, the method comprising administering to the mammal a composition comprising a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone species, and tryptanthrin.

In such a method of treating or inhibiting a pathological condition, the composition can contain a fraction isolated or derived from hops can be selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops. In another embodiment of the method, the fraction isolated or derived from hops comprises a compound of a supragenus having the formula:

T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, 1, and t orbital, with the proviso that if one of R, T, X, or Z is a it orbital, then the adjacent R, T, X, or Z is also a 'K orbital, thereby forming a double bond.

In the method, the fraction isolated or derived from hops can also comprise a compound of Genus A having the formula:

R"
00 o HO

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In yet another embodiment of the method, the fraction isolated or derived from hops can additionally comprise a compound of Genus B having the formula:

O o R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of the method, the fraction isolated or derived from hops comprises a compound selected from the group consisting of humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. In a particular embodiment, the composition can comprise about 0.5 to 10000 mg or about 50 to 7500 mg of the fraction isolated or derived from hops. Furthermore, the composition can comprise about 0.001 to 10 weight percent or about 0.1 to 1 weight percent of the fraction isolated or derived from hops. In a particular embodiment, the second component is rosemary. In another embodiment, the second component is an extract derived from rosemary. In still another embodiment, the second component is a triterpene species.
In a method of the invention, the composition can further comprise a third component different from the second component, where the third component is selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone species, and tryptanthrin. In a particular embodiment, the second and third components are an extract derived from rosemary and tryptanthrin, respectively.

In another embodiment of the method, the second component can be a compound derived from rosemary that is selected from the group consisting of 1,8-cineole, 19-alpha-hydroxyursolic acid, 2-13-hydroxyoleanolic acid, 3-0-acetyloleanolic acid, 3-0-acetylursolic acid, 6-methoxy-luteolin-7-glucoside, 6-methoxyluteolin, 6-methoxyluteolin-7-glucoside, methoxyluteolin-7-methylether, 7-ethoxy-rosmanol, methoxy-rosmanol, alpha-amyrin, alpha-humulene, alpha-hydroxyhydrocaffeic acid, alpha-pinene, alpha-terpinene, alpha-terpinenyl acetate, alpha-terpineol, alpha-thujone, apigenin, apigenin-7-glucoside, curcumene, benzyl-alcohol, B-amyrenone, B-amyrin, B-elemene, B-pinene, betulin, betulinic acid, borneol, bornyl-acetate, caffeic acid, camphene, camphor, carnosic acid, carnosol, carvacrol, carvone, caryophyllene, caryophyllene-oxide, chlorogenic acid, diosmetin, gamma-terpinene, hesperidin, isoborneol, limonene, luteolin, luteolin-3'-O-(3"-O-acetyl)-B-D-glucuronide, luteolin-3'-O-(4"-O-acetyl)-B-D-glucuronide, luteolin-3'-O-B-D-glucuronide, luteolin-7-glucoside, methyl-eugenol, myrcene, neo-chlorogenic acid, nepetin, octanoic acid, oleanolic acid, p-cymene, piperitenone, rosmanol, rosmaric acid, rosmaricine, rosmaridiphenol, rosemarinic acid, rosmarinol, rosmariquinone, sabinene, sabinyl acetate, salicylates, salicylic acid-2-B-D-glucoside, squalene, terpinen-4-ol, terpinolene, thymol, trans-anethole, trans-carveol, ursolic acid, verbenone, and zingiberene.

In such a method of the invention, the second component can also be a compound derived from rosemary that is selected from the group consisting of betulin, betulinic acid, carnosic acid, carnosol, carvacrol, chlorogenic acid, diosmetin, limonene, and luteolin. The composition used in the method can comprise about 0.5 to 5000 mg of the second component, or about 5 to 2000 mg of the second component, wherein the second component is selected from the group consisting of rosemary, extract derived from rosemary, and a compound derived from rosemary. In still another embodiment, the second component used in a method of the invention can be a triterpene species or a diterpene lactone species that is conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, acetate, and glutathione.

In yet another embodiment of a method of the invention, the second component can be a triterpene species that is selected from the group consisting of 18-a-glycyrrhetinic acid, 18-13-glycyrrhetinic acid, 2-a-3-a-dihydrooxyurs-12-3n-28-onic acid, 3-a-hydroxyursolic acid, 3-oxo-ursolic acid, betulin, betulinic acid, celastrol, eburicoic acid, friedelin, glycyrrhizin, gypsogenin, oleanolic acid, oleanolic acid-3-acetate, pachymic acid, pinicolic acid, sophoradiol, soyasapogenol A, soyasapogenol B, tripterin, triptophenolide, tumulosic acid, ursolic acid, ursolic acid-3-acetate, uvaol, and 13-sitosterol. In addition, the second component can be a triterpene species that is selected from the group consisting of 18-a-glycyrrhetinic acid, 18-B-glycyrrhetinic acid, 2-a-3-a-dihydrooxyurs-12-3n-28-onic acid, 3-a-hydroxyursolic acid, 3-oxo-ursolic acid, betulin, betulinic acid, celastrol, friedelin, oleanolic acid, tripterin, triptophenolide, ursolic acid, and uvaol.

In a particular embodiment of a method of the invention, the composition can comprise about 0.035 to 3500 mg of a triterpene species or about 0.7 to 700 mg of a triterpene species, wherein the second component is a triterpene species. In another embodiment of the method, the second component is tryptanthrin that is conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, acetate, and glutathione. In still another embodiment of a method of the invention, the composition can comprise about 0.035 to 3500 mg of tryptanthrin, or about 0.7 to 700 mg of tryptanthrin, wherein the second component is 5 tryptanthrin. In addition, the composition used in a method can comprise about 0.001 to 10 weight percent of the second component or about 0.1 to 1 weight percent of the second component. Furthermore, a ratio of the first component to the second component can be in the range of about 100:1 to about 1:100 or in the range of about 50:1 to about 1:50.

10 In such a method of treating or inhibiting a pathological condition, the pathological condition can be selected from the group consisting of autoimmune diseases, inflammatory diseases, neurological diseases, and cancer. In addition, the pathological condition can be selected from the group consisting of inflammation, inflammation-associated disorders, arthritis, asthma, bronchitis, menstrual cramps, 15 tendonitis, bursitis, skin-related conditions, gastrointestinal conditions, cancer, ophthalmic diseases, pulmonary inflammation, nervous system disorders, allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous damage. In methods of the invention, the composition can be administered in a variety of ways, including orally, topically, parenterally, or rectally.

20 The invention further provides a method of modulating the amount of cyclooxygenase-2 (COX-2) activity in target cells without substantially modulating COX-2 activity in non-target cells, the method comprising contacting the cells with a composition comprising a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from 25 rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone species, and tryptanthrin. In such a method of the invention, the non-target cells can also be contacted with a fraction isolated or derived from hops. The contacting step can be performed in vivo. In a method of the invention, the COX-2 activity can be modulated by inhibition of the COX-2 gene.

Additionally, the invention provides a method of treating or inhibiting a pathological condition in a mammal involving inhibiting inducibility or activity of cyclooxygenase-2 (COX-2), the method comprising administering to the mammal a composition comprising a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone, and tryptanthrin. In such a method of the invention, the fraction isolated or derived from hops can be selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

If desired in such a method of the invention, the fraction isolated or derived from hops comprises a compound of a supragenus having the formula:

R O O
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and 7t orbital, with the proviso that if one of R, T, X, or Z is a is orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In such a method of the invention, the fraction isolated or derived from hops can also comprise a compound of Genus A having the formula:

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In such a method of the invention, the fraction isolated or derived from hops can also comprise a compound of Genus B having the formula:

HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In such a method of the invention, the fraction isolated or derived from hops can comprise a compound selected from the group consisting of humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. In addition, the second component can be an extract derived from rosemary. Furthermore, the second component can be a triterpene species.

In another embodiment of a method of the invention, the composition further can comprise a third component different from the second component, the third component being selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone, and tryptanthrin. In a particular embodiment, the second and third components are an extract derived from rosemary and tryptanthrin, respectively.
In yet another embodiment of such a method of the invention, the second component is a compound derived from rosemary that is selected from the group consisting of 1,8-cineole, 19-alpha-hydroxyursolic acid, 2-B-hydroxyoleanolic acid, 3-O-acetyloleanolic acid, 3-0-acetylursolic acid, 6-methoxy-luteolin-7-glucoside, 6-methoxyluteolin, 6-methoxyluteolin-7-glucoside, methoxyluteolin-7-methylether, ethoxy-rosmanol, 7-methoxy-rosmanol, alpha-amyrin, alpha-humulene, alpha-hydroxyhydrocaffeic acid, alpha-pinene, alpha-terpinene, alpha-terpinenyl acetate, alpha-terpineol, alpha-thujone, apigenin, apigenin-7-glucoside, curcumene, benzyl-alcohol, B-amyrenone, B-amyrin, B-elemene, B-pinene, betulin, betulinic acid, borneol, bornyl-acetate, caffeic acid, camphene, camphor, carnosic acid, carnosol, carvacrol, carvone, caryophyllene, caryophyllene-oxide, chlorogenic acid, diosmetin, gamma-terpinene, hesperidin, isoborneol, limonene, luteolin, luteolin-3'-O-(3"-O-acetyl)-B-D-glucuronide, luteolin-3'-O-(4"-O-acetyl)-B-D-glucuronide, luteolin-3'-O-B-D-glucuronide, luteolin-7-glucoside, methyl-eugenol, myrcene, neo-chlorogenic acid, nepetin, octanoic acid, oleanolic acid, p-cymene, piperitenone, rosmanol, rosmaric acid, rosmaricine, rosmaridiphenol, rosemarinic acid, rosmarinol, rosmariquinone, sabinene, sabinyl acetate, salicylates, salicylic acid-2-B-D-glucoside, squalene, terpinen-4-ol, terpinolene, thymol, trans-anethole, trans-carveol, ursolic acid, verbenone, and zingiberene.

In another embodiment of such a method of the invention, the second component can be a triterpene species or a diterpene lactone species that is conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, acetate, and glutathione. Additionally, the second component can be a triterpene species that is selected from the group consisting of 18-a-glycyrrhetinic acid, 18-B-glycyrrhetinic acid, 2-a-3-a-dihydrooxyurs-12-3n-28-onic acid, 3-a-hydroxyursolic acid, 3-oxo-ursolic acid, betulin, betulinic acid, celastrol, eburicoic acid, friedelin, glycyrrhizin, gypsogenin, oleanolic acid, oleanolic acid-3-acetate, pachymic acid, pinicolic acid, sophoradiol, soyasapogenol A, soyasapogenol B, tripterin, triptophenolide, tumulosic acid, ursolic acid, ursolic acid-3-acetate, uvaol, and B-sitosterol. Also, the second component can be tryptanthrin that is conjugated to a member selected from the group consisting of mono- or di-saccharides, amino acids, sulfates, succinate, acetate, and glutathione. If desired in such a method of the invention, a ratio of the first component to the second component can be in the range of about 100:1 to about 1:100 or in the range of about 50:1 to about 1:50.

In a particular embodiment of a method of the invention, the pathological condition involving inhibiting inducibility or activity of COX-2 can be selected from the group consisting of inflammation, inflammation-associated disorders, arthritis, asthma, bronchitis, menstrual cramps, tendonitis, bursitis, skin-related conditions, gastrointestinal conditions, cancer, ophthalmic diseases, pulmonary inflammation, nervous system disorders, allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous damage.

Also, the invention provides a method of inhibiting prostaglandin synthesis selectively in target cells, the method comprising contacting the cells with a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone, and tryptanthrin. In such a method of the invention, the fraction isolated or derived from hops can be selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In addition in such a method of the invention, the fraction isolated or derived from hops can comprise a compound of a supragenus having the formula:

R O O

"
T HO R"
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, 5 CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and 't orbital, with the proviso that if one of R, T, X, or Z is a 7t orbital, then the adjacent R, T, X, or Z is also a 7t orbital, thereby forming a double bond.

10 In another embodiment of the method of the invention, the fraction isolated or derived from hops can comprise a compound of Genus A having the formula:

R..
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and 15 wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of such a method of the invention, the fraction isolated or derived from hops comprises a compound of Genus B having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In another embodiment of such a method of the invention, the fraction isolated or derived from hops can comprise a compound selected from the group consisting of humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

In a further embodiment, the invention provides a method of inhibiting an inflammatory response selectively in target cells, the method comprising contacting the cells with a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone, and tryptanthrin. In such a method, the fraction isolated or derived from hops can be selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In such a method, the fraction isolated or derived from hops can comprise a compound of a supragenus having the formula:

R"
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and 71 orbital, with the proviso that if one of R, T, X, or Z is a it orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In another embodiment of such a method, the fraction isolated or derived from hops can comprise a compound of Genus A having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of such a method, the fraction isolated or derived from hops can comprise a compound of Genus B having the formula:

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In such a method of the invention, the fraction isolated or derived from hops can comprise a compound selected from the group consisting of humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoaohumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

In yet another embodiment, the invention provides a method of modulating the inflammatory response in cells, the method comprising contacting the cells with a composition comprising a fraction isolated or derived from hops. In an additional embodiment, the invention provides a method of treating or inhibiting a pathological condition in a mammal associated with tissue-specific activation of inflammation, the method comprising administering to the mammal a composition comprising a fraction derived from hops. In such a method, the fraction derived from hops can be selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In an embodiment of such a method, the fraction derived from hops can comprise a compound of a supragenus having the formula:

R O O

T HO R"
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and 7c orbital, with the proviso that if one of R, T, X, or Z is a n orbital, then the adjacent R, T, X, or Z is also a 7C orbital, thereby forming a double bond.

In another embodiment of such a method of the invention, the fraction derived from hops can comprise a compound of Genus A having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In addition in such a method, the fraction derived from hops can comprise a compound of Genus B having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, 5 CH2CH(CH3)2, and CH(CH3)CH2CH3.

In another embodiment of such a method of the invention, the fraction derived from hops can comprise a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, 10 tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. In a particular embodiment of the method, the composition can comprise about 0.5 to 10000 mg or about 50 to 7500 mg of the fraction derived from hops. In addition, the composition can comprise about 0.001 to 10 weight percent or about 0.1 to 1 weight percent of the fraction 15 derived from hops.

In such a method of the invention, the pathological condition can be selected from the group consisting of autoimmune diseases, inflammatory diseases, neurological diseases, and cancer. In another embodiment, the pathological condition can be selected from the group consisting of inflammation, inflammation-associated 20 disorders, arthritis, asthma, bronchitis, menstrual cramps, tendonitis, bursitis, skin-related conditions, gastrointestinal conditions, cancer, ophthalmic diseases, pulmonary inflammation, nervous system disorders, allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous damage.

In still another embodiment, the invention provides a method of modulating the 25 amount of cyclooxygenase-2 (COX-2) activity in target cells without substantially modulating COX-2 activity in non-target cells, the method comprising contacting the cells with a fraction derived from hops. In such a method of the invention, the non-target cells can also be contacted with a fraction derived from hops. The contacting step can be performed in vivo. In the method of the invention, the COX-2 activity can be modulated by inhibition of COX-2 gene.

In yet another embodiment, the invention provides a method of treating or inhibiting a pathological condition in a mammal involving inhibiting inducibility or activity of cyclooxygenase-2 (COX-2), the method comprising administering to the mammal a composition comprising a fraction derived from hops. In such a method, the fraction derived from hops can be selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In another embodiment of the method, the fraction derived from hops can comprise a compound of a supragenus having the formula:

R O O
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and a orbital, with the proviso that if one of R, T, X, or Z is a 7t orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In such a method, the fraction derived from hops can comprise a compound of Genus A having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In another embodiment of the method, the fraction derived from hops can comprise a compound of Genus B having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of the method, the fraction derived from hops can comprise a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

In such a method of the invention, the pathological condition can be selected from the group consisting of wherein the pathological condition is selected from the group consisting of inflammation, inflammation-associated disorders, arthritis, asthma, bronchitis, menstrual cramps, tendonitis, bursitis, skin-related conditions, gastrointestinal conditions, cancer, ophthalmic diseases, pulmonary inflammation, nervous system disorders, allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous damage.

Moreover, the invention provides a method of inhibiting prostaglandin synthesis selectively in target cells, the method comprising contacting the cells with a fraction derived from hops. In such a method, the fraction derived from hops can be selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In another embodiment of the method, the fraction derived from hops can comprise a compound of a supragenus having the formula:

R O O
T HO
R' OH
X

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and it orbital, with the proviso that if one of R, T, X, or Z is a it orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In still another embodiment of the method, the fraction derived from hops comprises a compound of Genus A having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In yet another embodiment of the method, the fraction derived from hops can comprise a compound of Genus B having the formula:

O O
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In a further embodiment of the method, the fraction derived from hops can comprise a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

The invention further provides a method of modulating NF--KB in cells not associated with bone resorption, the method comprising contacting the cells with a composition comprising a fraction isolated or derived from hops. The invention additionally provides a method of treating or inhibiting a pathological condition other than osteoporosis in a mammal associated with tissue-specific activation of NF-KB, the method comprising administering to the mammal a composition comprising a 5 fraction isolated or derived from hops. In such a method, the fraction can be derived from hops and selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In another embodiment of the method, the fraction can be derived from hops and can comprise a compound of a supragenus having the formula:

T HO
R' OH
X

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and 15 wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and t orbital, with the proviso that if one of R, T, X, or Z is a orbital, then the adjacent R, T, X, or Z is also a t orbital, thereby forming a double bond.

In another embodiment of the method, the fraction is derived from hops and 20 comprises a compound of Genus A having the formula:

O o R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of the method, the fraction is derived from hops and comprises a compound of Genus B having the formula:

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In yet another embodiment of the method, the fraction is derived from hops and comprises a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

In a particular embodiment of the method, the composition used in a method of the invention can comprise about 0.5 to 10000 mg or about 50 to 7500 mg of the fraction isolated or derived from hops. Also, the composition can comprise about 0.00 1 to 10 weight percent or about 0.1 to 1 weight percent of the fraction isolated or derived from hops.

In a method of the invention modulating NFicB, the pathological condition can be selected from the group consisting of autoimmune diseases, inflammatory diseases, neurological diseases, cardiovascular diseases, and cancer. Also, the pathological condition in a method of modulating NFicB can be selected from the group consisting of asthma, HIV-1 replication, cold, and flu.

In another embodiment, the invention provides a method of modulating the amount of cyclooxygenase-2 (COX-2) activity in target cells not associated with bone resorption without substantially modulating COX-2 activity in non-target cells, the method comprising contacting the cells with a fraction isolated or derived from hops.
In the method, the non-target cells can also be contacted with a fraction isolated or derived from hops. The contacting step can be performed in vivo. In the method, the COX-2 activity can be modulated by inhibition of the COX-2 gene.
Additionally, the invention provides a method of treating or inhibiting a pathological condition other than osteoporosis in a mammal involving inhibiting inducibility or activity of cyclooxygenase-2 (COX-2), the method comprising administering to the mammal a composition comprising a fraction isolated or derived from hops. In a particular embodiment, the fraction can be derived from hops and selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In another embodiment of the method, the fraction is derived from hops and comprises a compound of a supragenus having the formula:

R O O
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and a orbital, with the proviso that if one of R, T, X, or Z is a it orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In still another embodiment of the method, the fraction is derived from hops and comprises a compound of Genus A having the formula:

o O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In yet another embodiment of the method, the fraction can be derived from hops and comprise a compound of Genus B having the formula:

O O
R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In a particular embodiment of the method, the fraction can be derived from hops and can comprise a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

In another embodiment of the method, the pathological condition can be selected from the group consisting of inflammation, inflammation-associated disorders, arthritis, asthma, bronchitis, menstrual cramps, tendonitis, bursitis, skin-related conditions, gastrointestinal conditions, cancer, ophthalmic diseases, pulmonary inflammation, nervous system disorders, allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, and central nervous damage.

Also, the invention provides a method of inhibiting prostaglandin synthesis selectively in target cells, the method comprising contacting the cells with a fraction derived from hops. In such a method, the fraction derived from hops can be selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In such a method, the fraction derived from hops can comprise a compound of a supragenus having the formula:

R O O
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and t orbital, with the proviso that if one of R, T, X, or Z is a t orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In another embodiment of the method, the fraction derived from hops can comprise a compound of Genus A having the formula:

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, 5 OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In yet another embodiment of the method, the fraction derived from hops comprises a compound of Genus B having the formula:

R..
HO
R' OH
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of the method, the fraction derived from hops can comprise a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.

Moreover, the invention provides a method of inhibiting an inflammatory response selectively in target cells, the method comprising contacting the cells with a fraction derived from hops. In such a method, the fraction derived from hops can be selected from the group consisting of isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.

In such a method, the fraction derived from hops can comprise a compound of a supragenus having the formula:

R O O
T HO
R' OH
X
Z
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl;
wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3; and wherein R, T, X, and Z are independently selected from the group consisting of H, F, Cl, Br, I, and it orbital, with the proviso that if one of R, T, X, or Z is a it orbital, then the adjacent R, T, X, or Z is also a it orbital, thereby forming a double bond.

In another embodiment of the method, the fraction derived from hops can comprise a compound of Genus A having the formula:

O O

R"
HO
R' OH

wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In still another embodiment of the method, the fraction derived from hops can comprise a compound of Genus B having the formula:

R"
HO
R' OH
wherein R' is selected from the group consisting of carbonyl, hydroxyl, OR, and OCOR, wherein R is alkyl; and wherein R" is selected from the group consisting of CH(CH3)2, CH2CH(CH3)2, and CH(CH3)CH2CH3.

In such a method of the invention, the fraction derived from hops can comprise a compound selected from the group consisting of cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. It is understood that compositions of the invention disclosed herein can be used in the various methods of the invention, as disclosed herein. , The invention additional provides a method of treating or inhibiting obesity in a mammal, the method comprising adminstering to the mammal a composition comprising a fraction isolated or derived from hops and a second component selected from the group consisting of rosemary, an extract derived from rosemary, a compound derived from rosemary, a triterpene species, a diterpene lactone, and tryptanthrin or other compositions of the invention, as disclosed herein.

As disclosed herein in Examples 1, 2 and 27, the AGS gastric mucosal cell line can function as a model system for determining potential gastrointestinal toxicity of anti-inflammatory agents. In AGS cells, COX-1 is expressed four times greater than COX-2. A lower inhibition of PGE2 in AGS cells is favorable because the AGS
cell line expresses more COX-1, which maintains mucosal homeostasis. The invention thus also provides a method of determining potential gastrointestinal toxicity of an anti-inflammatory agent. The method can include the steps of contacting an AGS
gastric mucosal cell with an anti-inflammatory agent; contacting a target inflammatory cell, for example, an A549 cell, with the anti-inflammatory agent; determining the 50% inhibitory concentration (IC50) of prostaglandin E2 (PGE2) expression for the anti-inflammatory agent in each of the AGS cell and target inflammatory cell;
and determining the ratio of the IC50 value of the AGS cell to the IC50 value of the target inflammatory cell, wherein a ratio greater than 1 indicates decreased potential gastrointestinal toxicity and a ratio less than 1 indicates increased potential gastrointestinal toxicity.

As disclosed herein, reduced isomerized alpha acids and isomerized alpha acids appear to inhibit COX-2 expression rather than directly on PGE2 (see Example 25).
Further as disclosed herein, hops has no significant dose-related effect on COX-1 or COX-2 enzyme activity, supporting that hops and/or fractions isolated or derived from hops affect COX-2 expression (see Example 26).

The description below is of specific examples setting forth preferred embodiments and are not intended to limit the scope.

AGS GASTRIC MUCOSAL CELLS CONSTITUTIVELY EXPRESS BOTH

Summary - This example demonstrates that the AGS human gastric mucosal cell line, possessing constitutive expression of COX-1 and COX-2, has excellent potential to serve as a model for assessing the gastrointestinal toxicity of cyclooxygenase-inhibiting compounds.

Equipment used in this example included: an OHAS Model #E01140 analytical balance, a Forma Model #F 1214 biosafety cabinet (Marietta, Ohio), various pipettes to deliver 0.1 to 100 1L (VWR, Rochester, NY), a cell hand tally counter (VWR
Catalog #23609-102, Rochester, NY), a Forma Model #F3210 CO2 incubator (Marietta, Ohio), a hemacytometer (Hausser Model #1492, Horsham, PA), a Leica Model #DM IL
inverted microscope (Wetzlar, Germany), a PURELAB Plus Water Polishing System (U.S. Filter, Lowell, MA), a 4 C refrigerator (Forma Model #F3775, Marietta, Ohio), a vortex mixer (VWR Catalog #33994-306, Rochester, NY), and a 37 C water bath (Shel Lab Model #1203, Cornelius, OR).

Chemicals and reagents - Prostaglandin E2 EIA kit Monoclonal was purchased from Cayman Chemical (Ann Arbor, MI). Anti-COX-1 and anti-COX-2 rabbit polyclonal antisera were obtained from Upstate Biotechnology (CITY, NY);
donkey anti-goat IgG-HRP was procured from Santa Cruz Biotechnology (City, CA). Heat inactivated Fetal Bovine Serum (FBS-HI Cat. #35-011CV), and Dulbeco's Modification of Eagle's Medium (DMEM Cat #10-013CV) was purchased from Mediatech (Herndon, VA). All standard reagents were obtained from Sigma (St.
Louis, MO) and were the purest commercially available.

Cell Culture - The human gastric mucosal cell line AGS was obtained from the American Type Culture Collection (ATCC number CRL-1739; Manassas, VA) and sub-cultured according to the instructions of the supplier. The cells were routinely cultured at 37 C with 5% CO2 in RPMI 1640 containing 10% FBS, with 50 units penicillin/mL, 50 gg streptomycin/mL, 5% sodium pyruvate, and 5% L-glutamine.
Exponentially growing cells were seeded into 6-well plates and grown to confluence.
A 20 gL aliquot of the supernatant media was sampled for determination of PGE2 content. Cells were then washed in PBS, scraped and lysed for immunoblotting.
Protein assay - Protein concentrations of cell lysates were determined using the NanoOrange Protein Quantitation Kit with bovine serum albumin as the standard (Molecular Probes, Eugene, OE) according to the procedure supplied by the manufacturer. Fluorescence was determined using a Packard FluoroCount, Model BF

10000 fluorometer with the excitation filter set at 485 nm and emission filter set at 570 nm using Packard PlateReader version 3.0 software. The I-Smart program provided with the Packard PlateReader was used to calculate the protein concentration.

5 Immunoblotting - Western blotting of COX-1 and COX-2 was performed using PAGErTM Gold Precast Gels (Bio Whittaker Molecular Applications (Rockland, ME). AGS cell lysates containing approximately 60 gg protein were loaded with Laemmli Sample Buffer into the wells of the gel in a total volume of 30 L.
The vertical minigel electrophoresis chambers were made by Savant Instruments Inc.
10 (Holbrook, NY), model MV 120. Gels were run at 40 mA/plate (constant current) at room temperature until the bromophenol blue stain reached the bottom of the gel, about one h. Gels were then blotted on the polyvinyl fluoride transfer membranes (Pall Corporation, Ann Arbor, MI), overnight, at 500 mA and 4 C. Precision Protein Standard molecular weight markers, unstained, broad range (BioRad, Hercules, CA) 15 were used. The BioWestTM Extended duration chemiluminescent substrate, a non-isotopic, horseradish peroxidase substrate kit for Western blot detection (Biolmaging Systems, Upland, CA) was used for protein visualization. Images of western blots were acquired using a UVP Epi Chemi II Darkroom (Biolmaging Systems), analyzed and enhanced by LabWorksTM Image Acquisition and Analysis Software (Biolmaging 20 Systems).

PGE2 assay - A commercial, non-radioactive procedure for quantification of was employed (Caymen Chemical, Ann Arbor, MI) and the recommended procedure of the manufacturer was used without modification. Briefly, 25 L of the medium, along with a serial dilution of PGE2 standard samples, were mixed with appropriate 25 amounts of acetylcholinesterase-labeled tracer and PGE2 antiserum, and incubated at room temperature for 18 h. After the wells were emptied and rinsed with wash buffer, 200 gL of Ellman's reagent containing substrate for acetylcholinesterase were added.
The reaction was carried out on a slow shaker at room temperature for 1 h and the absorbance at 415 nm was determined. The PGE2 concentration was represented as 30 picograms per 105 cells.

Results - As seen in Figure 6, the AGS cell line constitutively expresses both COX-1 and COX-2, with COX-1 expression approximately 4-times greater than COX-2 expression. PGE2 synthesis in AGS cells over 18 h was 660 pg/105 cells.
Thus, this example demonstrates that the AGS human gastric mucosal cell line, possessing constitutive expression of COX-1 and COX-2, has excellent potential to serve as a model for assessing the gastrointestinal toxicity of cyclooxygenase-inhibiting compounds.

In the past, the classical COX-2 hypothesis has downplayed the role of COX-2 expression in the gastrointestinal mucosa. While in normal gastric mucosa COX-1 is the predominant COX isozyme, as demonstrated in this example and in the literature, there is increasing evidence that detectable amount of COX-2 mRNA and protein are both constitutively expressed and inducible in specific locations of the gastric mucosa in both animals and humans [Halter, F., et al. (2001) Cyclooxygenase 2-implications on maintenance of gastric inucosal integrity and ulcer healing: controversial issues and perspectives. Gut 49, 443-453]. Recent studies in rats have shown that whereas selective inhibition of COX-1 or COX-2 is not ulcerogenic, combined inhibition of both COX-1 and COX-2 induces severe lesions in the stomach and small intestine comparable with the effects of NSAID such as indomethacin. This observation suggests an important contribution of COX-2 to the maintenance of gastrointestinal mucosal integrity.

NONSTEROIDAL ANTI-INFLAMMATORY DRUGS

Summary - This example illustrates that inhibition of PGE2 Synthesis in AGS
gastric cells by NSAIDs correlates with their observed clinical gastric irritation.
Chemicals - Rofecoxib and celexocib were obtained. Diisofluorophosphate (DIFP), nimensulide, ibuprofen, salicylic acid, aspirin, indomethacin and acetaminophen were purchased from Sigma (St. Louis, MO). All other chemicals were obtained from suppliers as described in Example 1.

Cells - A549 (human pulmonary epithelial; ATCC number CCL-185) and AGS
cells (human gastric mucosa; ATCC number CRL-1739) were obtained from the American Type Culture Collection (Manassas, VA) and sub-cultured according to the instructions of the supplier. The cells were routinely cultured at 37 C with 5% CO2 in RPMI 1640 containing 10% FBS, with 50 units penicillin/mL, 50 g streptomycin/mL, 5% sodium pyruvate, and 5% L-glutamine. On the day of the experiments, exponentially growing cells were harvested and washed with serum-free RPMI 1640.

The log phase A549 and AGS cells were plated at 8 x 104 cells per well in 0.2 mL
growth medium per well in a 96-well tissue culture plate. For the determination of PGE2 inhibition by the test compounds in A549 cells, the procedure of Warner et al., also known as the WHMA-COX-2 protocol [Warner, T. D., et al. (1999) Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci U S
A 96, 7563-7568.] was followed with no modifications. Briefly, 24 hours after plating of the A549 cells, interleukin-113 (10 ng/mL) was added to induce the expression of COX-2. After 24 hr, the cells were washed with serum-free RPMI 1640 and the test materials, dissolved in DMSO and serum-free RPMI, were added to the wells to achieve final concentrations of 25, 5.0, 0.5 and 0.05 g/mL. Each concentration was run in duplicate. DMSO was added to the control wells in an equal volume to that contained in the test wells. Sixty minutes later, A23187 (50 M) was added to the wells to release arachidonic acid. Twenty-five gL of media were sampled from the wells 30 minutes later for PGE2 determination.

Non-stimulated AGS cells were used in these studies. Twenty-four hours after plating in the 96-well microtiter plates, the cells were washed with serum-free RPMI
1640 and the test materials, dissolved in DMSO and serum-free RPM!, were added to the wells to achieve final concentrations of 25, 5.0, 0.5 and 0.05 g/mL. Each concentration was run in duplicate. DMSO was added to the control wells in an equal volume to that contained in the test wells. Sixty minutes later, arachidonic acid was added to the wells to achieve a final concentration of 100 M. Twenty-five gL
of media were sampled from the wells 30 minutes after the addition of arachidonic acid for PGE2 determination.

Cell viability - Cell viability was assessed by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (M"-based colorimetric assay (Sigma, St. Louis, MO). The MTT solution was added directly to the wells after sampling for PGE2 determination. The absorbance of each well was read at 580 nm using an ELISA
plate reader. No toxicity was observed at the highest concentrations tested for any of the compounds.

Calculations - The median inhibitory concentration (ICso) for PGE2 synthesis was calculated using CalcuSyn (BIOSOFT, Ferguson, MO). This statistical package performs multiple drug dose-effect calculations using the median effect methods described by T-C Chou and P. Talaly [(1984) Quantitative analysis of dose-effect.
relationships: the combined effects of multiple drugs or enzyme inhibitors.
Adv Enzyme Regul 22, 27-55.].

Briefly, the analysis correlates the "Dose" and the "Effect" in the simplest possible form: fa/fu = (C/Cm)m, where C is the concentration or dose of the compound and Cm is the median-effective dose signifying the potency. Cm is determined from the x-intercept of the median-effect plot. The fraction affected by the concentration of the test material is fa and the fraction unaffected by the concentration is fu (fu =1- fa).
The exponent m is the parameter signifying the sigmoidicity or shape of the dose-effect curve. It is estimated by the slope of the median-effect plot.

The median-effect plot is a graph of x= log(C) vs y = log(fa/fu) and is based on the logarithmic form of Chou's median-effect equation. The goodness of fit for the data to the median-effect equation is represented by the linear correlation coefficient r of the median-effect plot. Usually, the experimental data from enzyme or receptor systems have an r > 0.96, from tissue culture an r > 0.90 and from animal systems an r > 0.85. In the cell-based studies reported here, all linear correlation coefficients were greater than 0.90. Experiments were repeated three times on three different dates.

The percent inhibition at each dose was averaged over the three independent experiments and used to calculate the median inhibitory concentrations reported.

Results - The highly specific COX-2 inhibitor diisofluorophosphate exhibited a median inhibitory concentration in A549 cells of 1.19 g/mL and did not inhibit PGE2 synthesis in AGS cells at the highest concentration tested of 25 g/mL (Table 3).
Rofecoxib, and celexocib, selective COX-2 drugs, were 27-, and 14-times, respectively, more potent inhibitors of PGE2 synthesis in the target A549 cells than in the non-target AGS gastric mucosal cells. This finding demonstrates not only selectivity, but also target-tissue selectivity consistent with their low gastrointestinal toxicity. Nimensulide, another new, selective COX-2 inhibitor was equally as potent in the inhibition of PGE2 synthesis in both cell lines. The anti-inflammatory agent acetaminophen, purported to inhibit an unidentified isozyme of COX (COX-3) and having low gastrointestinal toxicity, inhibited PGE2 biosynthesis in A549 cells but had no effect on PGE2 synthesis in AGS gastric mucosal cells.

Alternatively and consistent with their demonstrated clinical gastric toxicity, ibuprofen, aspirin and indomethacin all exhibited more inhibition of PGE2 synthesis in the AGS cell line than in the target A549 cells. Salicylic acid, an anti-inflammatory agent that inhibits the expression of COX-2 with little gastric irritation, was inactive in both cell models.

Table 3 Median inhibitory concentrations for test compounds in the A549 and AGS cell lines.

Compound IC50 A549 IC5o AGS IC5o AGS / IC5o A549 [ g/ML] [gg/mL]

Diisofluorophosphate 1.19 >25 >21 Rofecoxib 0.081 2.21 27.3 Celexocib 0.004 0.055 13.8 Nimensulide 0.10 0.11 1.0 Ibuprofen 0.10 0.05 0.50 Aspirin 0.48 0.09 0.19 Indomethacin 0.033 0.002 0.002 Salicylic acid >25 >25 >1 Acetaminophen 0.607 >25 >41 These results validate the use of the AGS gastric mucosal cell line to evaluate potential gastrointestinal toxicity of anti-inflammatory agents capable of inhibiting the synthesis of PGE2. They also demonstrate cellular specificity in the action of COX-5 inhibiting compounds. A ratio of 1 for IC50 AGS/IC50 A549 indicates IC50s that are the same for both the AGS cell and A549 cells. If the ratio is higher than 1 for IC50 AGS/IC50 A549, then the inhibition of PGE2 is lower for the AGS cells. A lower inhibition of PGE2 in AGS cells is favorable because AGS cell line expresses more COX-1, which maintains mucosal homeostasis.

MURINE MACROPHAGES BY HOPS (Humulus lupulus) COMPOUNDS AND
DERVIATIVES

Summary - This example illustrates the potency of hops fractions and derivatives 15 to inhibit COX-2 synthesis of PGE2 preferentially over COX- 1 synthesis of PGE2 in the murine macrophage model.

Chemicals and reagents - Bacterial lipopolysaccharide (LPS; B E. coli 055:B5) was from Sigma (St. Louis, MO). Hops fractions (1) alpha hop (1% alpha acids;
AA), (2) aromahop OE (10% beta acids and 2% isomerized alpha acids, (3) isohop 20 (isomerized alpha acids; IAA), (4) beta acid solution (beta acids BA), (5) hexahop gold (hexahydro isomerized alpha acids; HHIAA), (6) redihop (reduced isomerized-alpha acids; RIAA), (7) tetrahop (tetrahydro-iso-alpha acids THIAA) and (8) spent hops were obtained from Betatech Hops Products (Washington, D.C., U.S.A.). The spent hops were extracted two times with equal volumes of absolute ethanol.
The 25 ethanol was removed by heating at 40 C until a only thick brown residue remained.
This residue was dissolved in DMSO for testing in RAW 264.7 cells. Unless otherwise noted, all standard reagents were obtained from Sigma (St. Louis, MO) and were the purest commercially available. All other chemicals and equipment were as described in Examples 1 and 2.

Cell culture - RAW 264.7 cells, obtained from American Type Culture Collection (Catalog #TIB-71, Manassas, VA), were grown in Dulbecco's Modification of Eagle's Medium (DMEM, Mediatech, Herndon, VA) and maintained in log phase. The DMEM growth medium was made by adding 50 mL of heat inactivated FBS and 5 mL of penicillin/streptomycin to a 500 mL bottle of DMEM and storing at 4 *C.
The growth medium was warmed to 37'C in water bath before use.

On day one of the experiment, the log phase RAW 264.7 cells were plated at 8 x 104 cells per well in 0.2 mL growth medium per well in a 96-well tissue culture plate in the morning. At the end of the day one (6 to 8 h post plating), 100 L of growth medium from each well were removed and replaced with 100 L fresh medium.

A 1.0 mg/mL stock solution of LPS, used to induce the expression of COX-2 in the RAW 264.7 cells, was prepared by dissolving 1.0 mg of LPS in 1 mL DMSO. It was vortexed until dissolved and stored at 4 C. Before use, it was melted at room temperature or in a 37 C water bath.

On day two of the experiment, test materials were prepared as 1000X stock in DMSO. In 1.7 mL microfuge tubes, 1 mL DMEM without FBS was added for test concentrations of 0.05, 0.10, 0.5, and 1.0 gg/mL. Two gL of the 1000X DMSO
stock of the test material was added to the 1 mL of medium without FBS. The tube contained the final concentration of the test material concentrated 2-fold and the tube placed in an incubator for 10 minutes to equilibrate to 37 C.

For COX-2 associated PGE2 synthesis, 100 gL of medium were removed from each well of the cell plates prepared on day one and replaced with 100 L of equilibrated 2X final concentration of the test compounds. Cells were then incubated for 90 minutes. Twenty L of LPS were added to each well of cells to be stimulated to achieve a final concentration of 1 g LPS/mL and the cells were incubated for 4 h.

The cells were further incubated with 5 M arachidonic acid for 15 minutes.
Twenty-five L of supernatant medium from each well was transferred to a clean microfuge tube for the determination of PGE2 released into the medium.

Following the LPS stimulation, the appearance of the cells was observed and viability was determined as described in Example 2. No toxicity was observed at the highest concentrations tested for any of the compounds. Twenty-five L of supernatant medium from each well was transferred to a clean microfuge tube for the determination of PGE2 released into the medium. PGE2 was determined and reported as previously described in Example 1.

For COX-1 associated PGE2 synthesis, 100 L of medium were removed from each well of the cell plates prepared on day one and replaced with 100 L of equilibrated 2X final concentration of the test compounds. Cells were then incubated for 90 minutes. Next, instead of LPS stimulation, the cells were incubated with 100 gM arachidonic acid for 15 minutes. Twenty-five gL of supernatant medium from each well was transferred to a clean microfuge tube for the determination of released into the medium. The appearance of the cells was observed and viability was determined as described in Example 2. No toxicity was observed at the highest concentrations tested for any of the compounds. Twenty-five L of supernatant medium from each well was transferred to a clean microfuge tube for the determination of PGE2 released into the medium. PGE2 was determined and reported as previously described in Example 1. The median inhibitory concentrations (IC50) for PGE2 synthesis from both COX-2 and COX-1 were calculated as described in Example 2.

Table 4. COX-2 and COX-1 inhibition in RAW 264.7 cells by hop fractions and derviatives Test Material COX-2 COX-1 COX-1/COX-2 [ g/mLl [ g/mL]
Alpha hop (AA) 0.21 6.2 30 Aromahop OE 1.6 4.1 2.6 Isohop (IAA) 0.13 18 144 Beta acids (BA) 0.54 29 54 Hexahop (HHIAA) 0.29 3.0 11 Redihop (RIAA) 0.34 29 87 Tetrahop (THIAA) 0.20 4.0 21 Spent hops (EtOH) 0.88 21 24 As seen in Table 4, all hops fractions and derivative selectively inhibited over COX-1 in this target macrophage model. This was a novel and unexpected finding. The extent of COX-2 selectivity for the hops derivatives IAA and RIAA, respectively, 144- and 87-fold, was unanticipated. Such high COX-2 selectivity combined with low median inhibitory concentrations, has not been previously reported for natural products from other sources.

HOPS COMPOUNDS AND DERIVATIVES ARE NOT DIRECT
CYCLOOXYGENASE ENZYME INHIBITORS

Summary - This example illustrates that hops compounds and derivatives do not inhibit PGE2 synthesis in A549 pulmonary epithelial cells at physiologically relevant concentrations when tested using the WHMA-COX-2 protocol.

Chemicals - Hops and hops derivatives used in this example were previously described in Example 3. All other chemicals were obtained from suppliers as described in Examples 1 and 2.

Cells - A549 (human pulmonary epithelial) Cells were obtained from the American Type Culture Collection (Manassas, VA) and sub-cultured according to the instructions of the supplier. The cells were routinely cultured at 37 C with 5% CO2 in RPMI 1640 containing 10% FBS, with 50 units penicillin/mL, 50 g streptomycin/mL, 5% sodium pyruvate, and 5% L-glutamine. On the day of the experiments, exponentially growing cells were harvested and washed with serum-free RPMI 1640.

Log phase A549 cells were plated at 8 x 104 cells per well with 0.2 mL growth medium per well in a 96-well tissue culture plate. For the determination of inhibition by the test compounds, the procedure of Warner et al. [(1999) Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo- oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci U S
A 96, 7563-7568], also known as the WHMA-COX-2 protocol was followed with no modification. Briefly, 24 hours after plating of the A549 cells, interleukin-113 (10 ng/mL) was added to induce the expression of COX-2. After 24 hr, the cells were washed with serum-free RPMI 1640 and the test materials, dissolved in DMSO and serum-free RPMI, were added to the wells to achieve final concentrations of 25, 5.0, 0.5 and 0.05 g/mL. Each concentration was run in duplicate. DMSO was added to the control wells in an equal volume to that contained in the test wells.
Sixty minutes later, A23187 (50 M) was added to the wells to release arachidonic acid.
Twenty-five L of media were sampled from the wells 30 minutes later for PGE2 determination.

Cell viability was assessed as previously described in Example 2. No toxicity was observed at the highest concentrations tested for any of the compounds. PGE2 in the supernatant medium was determined and reported as previously described in Example 1.

The median inhibitory concentration (IC50) for PGE2 synthesis was calculated as previously described in Example 2.

Results - At the doses tested, the experimental protocol failed to capture a median effective concentration of any of the hops extracts or derivatives. Since the protocol 5 requires the stimulation of COX-2 expression prior to the addition of the test-compounds, the likely answer to the failure of the test materials to inhibit synthesis is that their mechanism of action is to inhibit the expression of the COX-2 isozyme and not activity directly. While some direct inhibition can be observed using the WHMA-COX-2 protocol, this procedure is inappropriate in evaluating the anti-10 inflammatory properties of hops compounds or derivatives of hops compounds.

BY HOPS (Humulus lupulus) COMPOUNDS AND DERVIATIYES
Summary - This example illustrates the lack of PGE2 inhibition by hops fractions 15 and in the AGS human gastric mucosal cell line implying low gastric irritancy, potential of these compounds.

Chemicals and reagents were used as described in Example 3. AGS cells were grown and used for testing hops compounds and derivatives as described in Example 2. PGE2 was determined and reported as previously described in Example 1. The 20 median inhibitory concentrations (IC50) for PGE2 synthesis from AGS cells were calculated as described in Example 2.

Table 5. Lack of PGE2 inhibition in AGS gastric mucosal cells by hop fractions and derviatives Test Material IC5o AGS
mL
Alpha hop AA >25 Aromahop OE >25 Isohop IAA >25 Beta acids 03A) >25 Hexahop HHIAA >25 Redihop (RIAA) >25 Tetrahop THIAA >25 Spent hops EtOH >25 As seen in Table 5, all hops fractions and derivatives were unable to inhibit synthesis by 50% or more at the highest concentrations tested in the AGS
gastric mucosal cell line. Based on the anti-inflammatory potency exhibited by these fractions in target macrophages, this was a novel and unexpected finding.

COMPOUNDS FOUND IN ROSEMARY

Summary - This example illustrates the anti-inflammatory effect of rosemary extract and compounds commonly found in rosemary, carnosic acid, ursolic acid and oleanolic acid in target cells and the effect of rosemary extract and oleanolic acid on PGE2 synthesis in gastrointestinal cells.

Equipment used, chemicals, cell handing and calculation of median inhibitory concentrations were performed as previously described in Examples 1, 2 and 3.
Carnosic acid, ursolic acid and oleanolic acid were obtained from Sigma (St.
Louis, MO). The rosemary extract was a hexane extract obtained from selected leaves of Rosinarinus officinalis by mean (95% +/- 3% rosemary extract) that complied with US
regulation (21 CFR 101-22). It was determined by HPLC analysis that the extract contained a minimum of 11% phenolic diterpenes (consisting of carnosic acid, carnosol, methyl carnosate, rosemadial, rosemarinic acid), 4.9% min carnosic acid, and a minimum of 7.6% the sum of carnosol + carnosic acid. The carnosic acid was purchased from Sigma (St. Louis, MO) and the oleanolic acid (80%) was obtained from Sabinsa (121 Ethel Road West, Piscataway, NJ).

Table 6. PGE2 inhibition in RAW 264.7 and AGS cells by a rosemary extract, carnosic acid, ursolic acid, and oleanolic acid.
Test Material (COX-2/COX-1)t RAW 264.7 RAW or AGS COX-1/COX-2 [gg/mL] [gg/mL]
Rosemary extract (RAW/AGS) 0.51 4.0 7.8 Carnosic acid (RAW/RAW) 0.50 231 470 Ursolic acid (RAW/RAW) 1.91 33 17 Oleanolic acid (RAW/RAW) 1.15 19 17 Oleanolic acid (RAW/AGS) 1.15 5.0 4.3 'Indicates the cell lines used to estimate inhibitor effects, respectively, on.COX-2 or COX-1 synthesis of PGE2. In all cases, LPS-stimulated RAW 264.7 cells were used to determine median inhibitory concentrations of COX-2 mediated PGE2 synthesis.
For the estimation of the effects of test materials on COX-1-mediated synthesis, either non-stimulated RAW264.7 or non-stimulated AGS cells were used.

Results - All test materials exhibited potent inhibition of PGE2 synthesis in LPS-stimulated RAW 264.7 cells indicating inhibition of the COX-2 isozyme (Table 6).
Surprisingly, the rosemary extract was more potent than ursolic and oleanolic acids and equal to pure carnosic acid in potency with a median inhibitory concentration of 0.5 gg test material/mL medium. Since the rosemary extract contained only 11%
carnosic acid or derivative, the inference is that the interaction of the carnosic acid derivatives or the myriad of other compounds in the rosemary extract were acting in concert or synergistically to provide such a potent inhibition of COX-2.
Alternatively, one of the compounds previously identified in rosemary and listed earlier has extremely high potency for inhibiting COX-2 mediated synthesis of PGE2.

In non-stimulated RAW 264.7 cells, the pure compounds were relatively inactive exhibiting IC50 values of 231, 33 and 19 gg/mL, respectively, for carnosic, ursolic and oleanolic acids. This indicated a strong preference for COX-2 inhibition over for synthesis of PGE2 in the RAW 264.7 target cell model. This extent of COX
isozyme selectivity has never been reported in the literature and was an unexpected result. In the AGS gastric mucosal cell line, however, both the rosemary extract and oleanolic acid exhibited potent inhibition of PGE2 synthesis.

OLEANOLIC ACID AND URSOLIC ACID

Equipment used, chemicals, cell handing and calculation of median inhibitory concentrations were performed as previously described in Examples 1, 2 and 3.
The hops C02-extract was purchased from Hopunion, (Yakama, WA) and contained 30 to 60% alpha-acids and 15 to 45% beta-acids. Oleanolic and ursolic acids and were obtained from Sigma (St. Louis, MO) and were the highest purity commercially available (>98%).

Synergy of test components was quantified using the combination index (CI) parameter. The CI of Chou-Talaly is based on the multiple drug-effect and is derived from enzyme kinetic models (Chou, T.-C. and Talalay, P. (1977) A simple generalized equation for the analysis of multiple inhibitions of Michaelis-Menten kinetic systems.
J. Biol. Chem. 252:6438-6442). The equation determines only the additive effect rather than synergism or antagonism. However, we define synergism as a more than expected additive effect, and antagonism as a less than expected additive effect as proposed by Cho and Talalay Using the designation of CI = 1 as the additive effect, we obtain for mutually exclusive compounds that have the same mode of action or for mutually non-exclusive drugs that have totally independent modes of action the following relationships: CI < 1, = 1, and > 1 indicating synergism, additivity and antagonism, respectively.

Results- The 4:1 (C02-extract:triterpenoid) combination tested in RAW 264.7 cells exhibited potent synergy over the entire dose-response curve.
Combination indexes computed for both test materials at the IC5o, IC75 and IC9o are presented in Table 7. As described in this example, the synergy of these combinations covered a concentration range of 0.001 to 50 gg/mL of each component of the combination.

Table 7. Computed Combination Indexes for the dose-response curves of 1:4 combinations of a C02-extract of hops and the triterpenes oleanolic and ursolic acid Test Material C150 CI75 090 Mean CI
C02-Extract:Oleanolic acid [1:4] 0.514 0.461 0.414 0.463 C02-Extract:Ursolic acid [1:4] 0.529 0.650 0.806 0.662 WITH AN EXTRACT OF ROSEMARY IN TARGET AND NONTARGET CELLS
Summary - This example illustrates synergy of combinations of reduced isomerized alpha acids and rosemary extract on target A549 cells and synergistic antagonism of rosemary inhibition of PGE2 synthesis in AGS gastric mucosal cells.
Equipment used, chemicals, cell handing and calculation of median inhibitory concentrations were performed as previously described in Examples 1, 2, 3 and 4.
Several differences in the protocol for testing in the A549 cells were incorporated in this example. First, test materials were added to the medium 60 minutes prior to stimulation with IL-1B. Second, in the determination of dose-response curves, arachidonic acid was used in place of the calcium ionophore A23187. Synergy of the combinations was computed as described in Example 7.

Results - Table 8 shows PGE2 inhibition by reduced isomerized alpha-acids, rosemary extract and a 2:1 combinations of reduced isomerized alpha-acids and rosemary extract in IL-113 stimulated A549 cells. This cell line represents a model target cell for anti-inflammatory efficacy. Median inhibitory concentrations for reduced isomerized alpha-acids and rosemary extract independently were, respectively, 0.84 and 1.3 g/mL. The 2:1 combination of-reduced isomerized alpha-acids and rosemary extract exhibited synergy at and below the median inhibitory concentration of the combination.

Table 9 shows inhibition of PGE2 synthesis in the human gastric AGS cells.
These cells represent a model for gastrointestinal toxicity of prostaglandin inhibitors.
Test materials exhibiting inhibition of PGE2 synthesis in these cells would be expected to demonstrate gastric irritation and ulceration with chronic use.
The 5 inhibition of PGE2 synthesis by rosemary extract was synergistically antagonized by a 2:1 combination of reduced isomerized alpha-acids and rosemary extract. This unexpected result represents a novel finding of synergistic antagonism.

Table 8. Median inhibitory concentrations and combination index for PGE2 inhibition by reduced isomerized alpha-acids, rosemary extract and a combination of isomerized 10 alpha-acids and rosemary extract in IL-113-stimulated A549 cells Test Material IC50 Combination Index <
[1g/ml] 1.0f [ g/mL]
Reduced isomerized alpha-acids (RIAA) 0.84 Rosemary extract 1.3 RIAA:Rosemary 2:1 0.48 At 0.48 and below f The combination index was less than 1 over the portion of the dose-response curve at and below the IC50 value indicating synergistic inhibition of PGE2 synthesis by the combination at these concentrations.

Table 9. Synergy of a 1:1 combination of reduced isomerized alpha-acids with 15 rosemary extract resulting in a reduction of PGE2 inhibition in AGS gastric mucosal cells.

Test Material IC50 Combination Index [ g/ml]
Reduced isomerized alpha-acids (RIAA) >25 -Rosemary 4.0 -RIAA:Rosemary 1:1 >25 >1.0f ]'The combination index was greater than 1 over the entire dose-response curve indicating synergistic antagonism of PGE2 inhibition by the combination.

While this example only presents the combination of rosemary extract with one of 20 the hops derivative, reduced isomerized alpha-acids, it would be obvious for one skilled in the art to assume to expect the same results with other hops derivatives that also show no PGE2 inhibition with AGS cells at dose as high as 25 gg/mL.
Examples of these hops derivatives would include isomerized-alpha acids, hexahydro-isomerized alpha acids, tetrahydro-iso-alpha acids and extracts of spent hops.

SYNERGISTIC INHIBITION OF PGE2, SYNTHESIS BY REDUCED ISOMERIZED
ALPHA-ACIDS AND OLEANOLIC ACID IN TARGET CELLS WITH NO

Summary - This example illustrates that reduced isomerized alpha-acids exhibit strong synergy with the triterpene oleanolic acid in the inhibition of PGE2 synthesis is the target A549 cells and synergistically antagonize oleanolic acid inhibition of PGE2 synthesis in gastric cells.

Equipment used, chemicals, cell handing and calculation of median inhibitory concentrations were performed as previously described in Examples 1, 2, 3 and 4.
Several differences in the protocol for testing in the A549 cells were incorporated in this example. First, test materials were added to the medium 60 minutes prior to stimulation with IL-1 B. Second, in the determination of dose-response curves, cells remained in the presence of test material overnight before the sampling of media for PGE2 determination. Synergy of the combinations was computed as described in Example 7. Reduced isomerized alpha-acids were obtained as a one percent aqueous solution from John Haas, Inc. (Yakima, WA) and oleanolic acid was obtained from Sabinsa (Piscataway, NJ) and was 80% pure. Synergy of the combinations was computed as described in Example 7.

Results - Table 10 shows PGE2 inhibition by oleanolic acid, reduced isomerized alpha-acids and various combinations of reduced isomerized alpha-acids and oleanolic acid in A549 cells. This cell line represents a model target cell for anti-inflammatory efficacy. Median inhibitory concentrations for reduced isomerized alpha-acids and oleanolic acid independently were, respectively, 0.03 and 0.39 gg/mL.
Combinations of reduced isomerized alpha-acids and oleanolic acid consisting of 10:1, 5:1, and 1:5, respectively, exhibited synergy on the dose-response curve at combined concentrations of 0.11, 0.38 and 0.76 gg/mL. Thus, when the sum of the two components was equal to or less than 0.11, 0.38 or 0.76 gg/mL, their ability to inhibit PGE2 synthesis was greater than the sum of their individual activities.

Table 10. Median inhibitory concentrations and combination indexes for PGE2 inhibition by reduced isomerized alpha-acids, oleanolic acid and four combinations of isomerized alpha-acids and oleanolic acid in IL-lB-stimulated A549 cells.

Test Material IC50 Combination Index < 1.0 [ g/ML]
Oeeanolic acid (80% Sabinsa) 0.390 -Reduced isomerized alpha-acids (RIAA) 0.028 -RIAA:Oleanolic acid - [10:1] 0.042 At 0.11 gg/mL and below RIAA:Oleanolic acid - [5:1] 0.059 At 0.38 gg/mL and below RIAA:Oleanolic acid - [1:5] 0.022 At 0.76 g/mL and below RIAA:Oleanolic acid - [1:10] 0.166 No ]' The combination index was less than 1 over the portion of the dose-response curve at the tabulated values indicating synergistic inhibition of PGE2 synthesis by the combination at and below these concentrations.

Table 11 shows inhibition of PGE2 synthesis in the human gastric AGS cells.
These cells represent a model for gastrointestinal toxicity of prostaglandin inhibitors.
Test materials exhibiting inhibition of PGE2 synthesis in these cells would be expected to demonstrate gastric irritation and ulceration with chronic use.
The inhibition of PGE2 synthesis by oleanolic acid was synergistically antagonized by all combinations with reduced isomerized alpha-acids. This unexpected result represents a novel finding of synergistic antagonism.

Table 11. Synergy of reduced isomerized alpha-acids with oleanolic acid resulting in a reduction of PGE2 inhibition in AGS gastric mucosal cells Test Material [ g/mL] Combination Index >1.Ot Oleanolic acid 5.0 -Reduced isomerized alpha-acids (RIAA > 25 -RIAA:Oleanolic acid - [10:1] > 25 Antagonism RIAA:Oleanolic acid - [5:1] > 25 Antagonism RIAA:Oleanolic acid - [1:5] > 25 Antagonism RIAA:Oleanolic acid - [1:10] > 25 Antagonism ]'When CI>1.0 at the IC50, the combination is said to exhibit antagonism in the inhibition of PGE2 synthesis by AGS cells.

While this example only presents the combination of oleanolic acid with one of the hops derivative, reduced isomerized alpha-acids, it would be obvious for one skilled in the art to assume to expect the same results with other hops derivatives that also show no PGE2 inhibition with AGS cells at dose as high as 25 g/mL.
Examples of these hops derivatives would include isomerized-alpha acids, hexahydro-isomerized alpha acids, tetrahydro-iso-alpha acids and extracts of spent hops.

REDUCED ISOMERIZED ALPHA ACIDS WITH TRYPTANTHRIN IN TARGET

Summary - This example illustrates a potent synergy of a 1:1 combination of reduced isomerized alpha acids and tryptanthrin on target A549 cells and synergistic antagonism of tryptanthrin inhibition of PGE2 synthesis in AGS gastric mucosal cells.

Equipment used, chemicals, cell handing and calculation of median inhibitory concentrations were performed as previously described in Examples 1, 2, 3, 4 and 9.
Reduced isomerized alpha-acids were obtained as a one percent aqueous solution from John Haas, Inc. (Yakima, WA) and tryptanthrin was obtained from Waco Chemicals (Richmond, VA) and was the highest purity commercially available. Several differences in the protocol for testing in the A549 cells were incorporated in this example. First, test materials were added to the medium 60 minutes prior to stimulation with IL-lB. Second, in the determination of dose-response curves, cells remained in the presence of test material overnight before the sampling of media for PGE2 determination. Synergy of the combinations was computed as described in Example 7.

Results - Table 12 shows PGE2 inhibition by reduced isomerized alpha-acids, tryptanthrin and a 1:1 combination of reduced isomerized alpha-acids and tryptanthrin in IL-1B stimulated A549 cells. This cell line represents a model target cell for anti-inflammatory efficacy. Median inhibitory concentrations for reduced isomerized alpha-acids and tryptanthrin independently were, respectively, 0Ø028 and 0.30 gg/mL. The 1:1 combination of reduced isomerized alpha-acids and tryptanthrin exhibited synergy over the entire dose-response curve.

Table 13 shows inhibition of PGE2 synthesis in the human gastric AGS cells.
These cells represent a model for gastrointestinal toxicity of prostaglandin inhibitors.
Test materials exhibiting inhibition of PGE2 synthesis in these cells would be expected to demonstrate gastric irritation and ulceration with chronic use.
The inhibition of PGE2 synthesis by tryptanthin was synergistically antagonized by a 1:1 combination of reduced isomerized alpha-acids and tryptanthrin or conjugates thereof.
This unexpected result represents a novel finding of synergistic antagonism.

Table 12. Median inhibitory concentrations and combination index for PGE2 inhibition by reduced isomerized alpha-acids, tryptanthrin and a combination of isomerized alpha-acids and tryptanthrin in IL-1B-stimulated A549 cells Test Material IC50 Combination Index [ g/mL]
Reduced isomerized alpha-acids RIAA 0.028 -Tryptanthrin 0.300 -RIA.A:Tryptanthrin - [1:1] 3.1 x 10"7 <1.O f ]' The combination index was less than 1 over the entire dose-response curve indicating synergistic inhibition of PGE2 synthesis by the combination.

Table 13. Synergy of combinations of reduced isomerized alpha-acids with tryptanthrin resulting in a reduction of PGE2 inhibition in AGS gastric mucosal cells.

Test Material [ g/mL] Combination Index Reduced isomerized alpha-acids (RIAA) > 25 -Tryptanthrin 4.2 -RIAA:Tryptanthrin - [1:1] >25 >1.0t -[The combination index was greater than 1 over the entire dose-response curve indicating synergistic antagonism of PGE2 inhibition by the combination.

While this example only presents the combination of trypanthrin with one of the hops derivative, reduced isomerized alpha-acids, it would be obvious for one skilled in the art to assume to expect the same results with other hops derivatives that also show no PGE2 inhibition with AGS cells at dose as high as 25 gg/mL. Examples of these hops derivatives would include isomerized-alpha acids, hexahydro-isomerized alpha acids, tetrahydro-iso-alpha acids and extracts of spent hops.

HUMAN RECEIVING A COMBINATION CONTAINING HOPS DERIVATIVES, A ROSEMARY EXTRACT AND OLEANOLIC ACID

Summary - This example demonstrates the presence of PGE2 inhibiting materials in a human subject following ingestion of a 5:5:1 combination of reduced isomerized alpha acids, rosemary extract and oleanolic acid three times per day for five days.

Equipment used, chemicals, RAW 264.7 cell handing and calculation of PGE2 concentrations were performed as previously described in Examples 1, 2, and 3.
Reduced isomerized alpha acids, rosemary extract and oleanolic acid were as described in Examples 3, 6 and 7, respectively. Gel caps were made to contain mg reduced isomerized alpha acids, 200 mg rosemary and 40 mg oleanolic acid in an oil base. Plasma samples were obtained from a human volunteer prior to and five days after consuming three capsules per day for five days. Capsules were taken at approximately eight-hour intervals throughout the day. On the fifth day, blood was drawn one hour before taking the last capsule and 1, 2, 4 and 7 hours after dosing. All PGE2 assays in plasma samples were replicated eight times. Outliers were defined and eliminated if the value was more than three standard deviations from the group mean computed without the perceived outlier. Raw data with and without the outliers were graphed. Concentrations of test material in plasma relating to percent inhibition were estimated using a standard curve of the combination in commercial plasma (Gibco, Grand Island, NY).

Figure 7[A] illustrates the inhibition of PGE2 synthesis by the plasma samples at the indicated times. A 9- to 3-fold increase in PGE2 inhibition was observed during the first post-dosing hour. Effective half-life (time to reduce the ability to inhibit PGE2 synthesis by one-half) of the test material was approximately four hours.

Estimates of test material relating to the observed percentage inhibition of synthesis in RAW 264.7 cells are presented in Figure 7[B]. Using only the data with outliers removed, a 12.5-fold increase in test material concentration was noted during the first hour. A maximal concentration of 880 ng/mL plasma was seen at both the 1 and 2 post-dosing hours. The concentration half-live was approximately 2.2 hours.
The lack of consistency between the effective half-life and concentration half-life may be inferred to be due to the synergy of components in the formulation.
Efficacy is extended due to positive and synergistic interactions among the isomerized alpha acids, the myriad of compounds in the rosemary extract and oleanolic acid as has been demonstrated by the examples in this application.

NORMALIZATION OF JOINT FUNCTION FOLLOWING TRAUMA
A representative composition of the preferred embodiments as a dietary supplement would be in an oral formulation, i.e. tablets or gel caps that would supply one of the following combinations: 0.1 to 10 mg isocohumulone/kg per day; 0.01 to 10 mg dihydroadhumulone/kg per day; 0.01 to 10 mg tetrahydro-isocohumulone/kg per day; 0.01 to 10 mg/kg per day of hexahydro-isohumulone/kg per day for a 70 kg person.

Normalization of joint movement following physical trauma due to exercise or repetitive movement stress would be expected to occur following two to ten doses.
This result would be expected in all animals.

NORMALIZATION OF JOINT FUNCTION FOLLOWING TRAUMA
A representative composition of the preferred embodiments as a dietary supplement would be in an oral formulation, i.e. tablets or gel caps that would supply one of the following combinations:

17 mg reduced isomerized alpha-acid/kg per day, 17 mg rosemary extract/kg per day and 17 mg ursolic acid/kg per day;
17 mg reduced isomerized alpha-acid/kg per day, 17 mg rosemary extract/kg per day and 3.4 mg ursolic acid/kg per day;
34 mg reduced isomerized alpha-acid/kg per day, 34 mg rosemary extract/kg per day and 3.4 mg ursolic acid/kg per day;
340 mg reduced isomerized alpha-acid/kg per day, 340 mg rosemary extract/kg per day and 3.4 mg ursolic acid/kg per day;
17 mg reduced isomerized alpha-acid/kg per day, 17 mg rosemary extract/kg per day and 85 mg ursolic acid/kg per day;
17 mg reduced isomerized alpha-acid/kg per day, 17 mg rosemary extract/kg per day and 170 mg ursolic acid/kg per day; or 17 mg reduced isomerized alpha-acid/kg per day, 17 mg rosemary extract/kg per day and 1700 mg ursolic acid/kg per day for a 70 kg person.

Normalization of joint movement following physical trauma due to exercise or repetitive movement stress would be expected to occur following two to ten doses.
This result would be expected in all animals.

CLINICAL EFFECTIVENESS OF LOTION FORMULATIONS IN THE
TREATMENT OF ACNE ROSACEA

A lotion designed to contain one of the following:

1. 0.1% wt of the isomerized alpha-acid isocohumulone;

2. 0.1 % wt of the reduced isomerized alpha-acid dihydro-adhumulone;
3. 0.1% wt of the tetrahydroisoalpha-acid tetrahydro-isocohumulone ;
or 4. 0.1 % wt hexahydro-isohumulone is applied to affected areas of patients who have exhibited acne rosacea as diagnosed by their health practitioner and confirmed by an independent board-certified dermatologist.

Self-evaluation tests and are administered one week prior to the study to quantify the surface area affected and redness. In addition, similar variables are scored by the professional clinical staff not aware of the patients treatment status. These evaluations are repeated on Days 0, 7, 14 and 21.

Patients are randomly assigned to the test formulation or placebo at the start of the study. The test formulation and placebo are applied to the affected area one or two times per day. Treatment for health conditions such as diabetes, hypertension, etc. is allowed during the study. Scores are statistically compared between the test formulation and the placebo for each of the four observational periods.
Patients treated with the composition of the preferred embodiments in a lotion formulation are considered improved if the patients' scores improve by greater than 20% from the pre-test scores within each category evaluated. The percentage of persons exhibiting improvement is compared between the combination formulations and the placebo control. The difference between the two groups is considered statistically significant if the probability of rejecting the null hypothesis when true is less than five percent.

CLINICAL EFFECTIVENESS OF LOTION FORMULATIONS IN THE
TREATMENT OF ACNE ROSACEA

A lotion designed to contain one of the following:
1. 0.1% wt of the alpha-acid humulone;

2. 0.1% wt of the isomerized alpha-acid isocohumulone;

3. 0.1% wt of the reduced isomerized alpha-acid dihydro-adhumulone;
4. 0.1% wt of the tetrahydroisoalpha-acid tetrahydro-isocohumulone; or 5. 0.1 % wt of the hexahydroisoalpha-acid hexahydro-isohumulone is applied to affected areas of patients who have exhibited acne rosacea as diagnosed by their health practitioner and confirmed by an independent board-certified dermatologist.

Self-evaluation tests and are administered one week prior to the study to quantify the surface area affected and redness. In addition, similar variables are scored by the professional clinical staff not aware of the patients treatment status. These evaluations are repeated on Days 0, 7, 14 and 21.

Patients are randomly assigned to the test formulation or placebo at the start of the study. The test formulation and placebo are applied to the affected area one or two times per day. Treatment for health conditions such as diabetes, hypertension, etc. is allowed during the study. Scores are statistically compared between the test formulation and the placebo for each of the four observational periods.
Patients treated with the composition of the preferred embodiments in a lotion formulation are considered improved if the patients' scores improve by greater than 20% from the pre-test scores within each category evaluated. The percentage of persons exhibiting improvement is compared between the combination formulations and the placebo control. The difference between the two groups is considered statistically significant if the probability of rejecting the null hypothesis when true is less than five percent.

CLINICAL EFFECTIVENESS OF LOTION FORMULATIONS IN THE
TREATMENT OF ACNE ROSACEA

A lotion designed to contain one of the following:

1. 0.1 % wt of the alpha-acid humulone and 0.1 % trypanthrin;

2. 0.1% wt of the isomerized alpha-acid isocohumulone and 0.1%
trypanthrin;

3. 0.1 % wt of the reduced isomerized alpha-acid dihydro-adhumulone and 0.1 % tryptanthrin;

4. 0.1% wt of the tetrahydroisoalpha-acid tetrahydro-isocohumulone and 0.1 % tryptanthrin; or 5. 0.1 % wt of the hexahydroisoalpha-acid hexahydro-isohumulone and0.1 % tryptanthrin is applied to affected areas of patients who have exhibited acne rosacea as diagnosed by their health practitioner and confirmed by an independent board-certified dermatologist.

Self-evaluation tests and are administered one week prior to the study to quantify the surface area affected and redness. In addition, similar variables are scored by the professional clinical staff not aware of the patients treatment status. These evaluations are repeated on Days 0, 7, 14 and 21.

Patients are randomly assigned to the test formulation or placebo at the start of the study. The test formulation and placebo are applied to the affected area one or two times per day. Treatment for health conditions such as diabetes, hypertension, etc. is allowed during the study. Scores are statistically compared between the test formulation and the placebo for each of the four observational periods.
Patients treated with the composition of the preferred embodiments in a lotion formulation are considered improved if the patients' scores improve by greater than 20% from the pre-test scores within each category evaluated. The percentage of persons exhibiting improvement is compared between the combination formulations and the placebo control. The difference between the two groups is considered statistically significant if the probability of rejecting the null hypothesis when true is less than five percent.

CLINICAL EFFECTIVENESS OF A LOTION FORMULATION IN THE
TREATMENT OF PSORIASIS

This example is performed in the same manner as described in Examples 14, 15 and 16 except that the composition is applied to affected areas of patients who have exhibited psoriasis as diagnosed by their own practitioner and confirmed by an independent board-certified dermatologist. Self-evaluation tests are administered one week prior to the study to quantify the surface area affected and skin condition. In addition, similar variables are scored by the professional clinical staff not aware of the patients treatment status. These evaluations are repeated on Days 0, 7, 30 and 60.

Patients are randomly assigned to the test formulation or placebo at the start of the study. The test formulation and placebo are applied to the affected area one or two times per day. Treatment for health conditions such as diabetes, hypertension, etc. is allowed during the study. Scores are statistically compared between the test formulation and the placebo for each of the four observational periods.
Patients treated with the composition of the preferred embodiments as the test lotion formulation are considered improved if the patients' scores improve by greater than 20% from the pre-test scores within each category evaluated. The percentage of persons exhibiting improvement is compared between the test formulation and the placebo control.
The difference between the two groups is considered statistically significant if the probability of rejecting the null hypothesis when true is less than five percent.

CLINICAL EFFECTIVENESS OF A FORMULATION IN THE TREATMENT OF
ALZHEIMER'S DISEASE

An oral formulation as described in Examples 12 and 13 is administered to patients who have manifested an early stage of Alzheimer's Disease (AD), as diagnosed by their practitioner and confirmed by an independent board-certified neurologist. Two weeks before the clinical trial, the patients undergo appropriate psychoneurological tests such as the Mini Mental Status Exam (MMSE), the Alzheimer Disease Assessment Scale (ADAS), the Boston Naming Test (BNT), and the Token Test (TT). Neuropsychological tests are repeated on Day 0, 6 weeks and 3 months of the clinical trial. The tests are performed by neuropsychologists who are not aware of the patient's treatment regimen.

Patients are randomly assigned to the test formulation or placebo at the start of the study. The test formulation and placebo are taken orally one or two times per day.
Treatment for conditions such as diabetes, hypertension, etc. is allowed during the study. Scores are statistically compared between the test formulation and the placebo for each of the three observational periods. Without treatment, the natural course of AD is significant deterioration in the test scores during the course of the clinical trial.
Patients treated with the composition of the preferred embodiments as the test formulation are considered improved if the patients' scores remain the same or improve during the course of the clinical trial.

ORAL FORMULATION IN THE TREATMENT AND PREVENTION OF COLON
CANCER
An oral formulation as described in Examples 12 and 13 is administered to patients who have manifested an early stage of colon cancer as diagnosed by their own practitioner and confirmed by a independent board-certified oncologist.

Patients are randomly assigned to the test formulation or a placebo at the start of the study. The test formulation and placebo are taken orally one or two times per day.
Treatment for conditions such as diabetes, hypertension, etc. is allowed during the study. Endoscopic evaluations are made at one, two, six and twelve months.
Evidence of reappearance of the tumor during any one of the four follow-up clinical visits is considered a treatment failure. The percentage of treatment failures is compared between the test formulation and the placebo control. Under the experimental conditions described, the test material is expected to decrease the tumor incidence with respect to the control group. The difference between the two groups is considered statistically significant if the probability of rejecting the null hypothesis when true is less than five percent.

ORAL FORMULATION FOR THE TREATMENT OF IRRITABLE BOWEL
SYNDROME
An oral formulation as described in Examples 12 and 13 is administered to patients who have manifested irritable bowel syndrome as diagnosed by their practitioner. Normal bowel functioning is restored within 48 hours.

NORMALIZATION OF JOINT FUNCTIONING IN OSTEOARTHRITIS
Using compositions described in Examples 12 and 13 normalization of joint stiffness due to osteoarthritis occurs following five to twenty doses, in the presence or absence of glucosamine or chondroitin sulfate. In addition, the composition does not interfere with the normal joint rebuilding effects of these two proteoglycan constituents, unlike traditional non-steroidal anti-inflammatory agents.

PULMONARY CELLS

Summary - This example illustrates that house mite dust allergens can induce PGE2 biosynthesis in pulmonary epithelial cells.

Background Sensitivity to allergens is a problem for an increasing number of consumers.
This issue has been complicated by a surprising increase in asthma over the past few years.
Asthma suffers are especially sensitive to airborne allergens. Allergy rates are also on the rise. This gives rise to increased awareness of the causes of allergy symptoms and how to decrease the associated discomfort. Approximately 10% of the population become hypersensitized (allergic) upon exposure to antigens from a variety of environmental sources. Those antigens that induce immediate and/or delayed types of hypersensitivity are known as allergens. These include products of grasses, trees, weeds, animal dander, insects, food, drugs, and chemicals. Genetic predisposition of an individual is believed to play a role in the development of immediate allergic responses such as atopy and anaphylaxis whose symptoms include hay fever, asthma, and hives.

Many allergens are protein-based molecules, and these protein allergens can originate from many sources. It has been know for some time that one of the most common sources of allergens in a house is from dust mites. Of course, as is the case with all allergens, only certain people are allergic to dust mite allergens.
But this group of people can be quite large in many areas, especially in hot humid areas. For example, in the southeastern United States of America, where it is both hot and humid for much of the year, the incidence of house dust mite allergies in the general population can be as high as 25%. House dust mites thrive in plush carpets, overstuffed upholstery, cushy bed comforters and the like.

Methods Mite dust allergen isolation - Dermatophagoides farinae are the American house dust mite. D. farinae were cultured on a 1:1 ratio of Purina Laboratory Chow (Ralston Purina, Co, St. Louis, MO) and Fleischmann's granulated dry yeast (Standard Brands, Inc. New York, NY) at room temperature and 75% humidity. Live mites were aspirated from the culture container as they migrated from the medium, killed by freezing, desiccated and stored at 0% humidity. The allergenic component of the mite dust was extracted with water at ambient temperature. Five-hundred mg of mite powder were added to 5 mL of water (1:10 w/v) in a 15 mL conical centrifuge tube (VWR, Rochester, NY), shaken for one minute and allowed to stand overnight at ambient temperature. The next day, the aqueous phase was filtered using a 0.2 m disposable syringe filter (Nalgene, Rochester, NY). The filtrate was termed mite dust allergen and used to test for induction of PGE2 biosynthesis in A549 pulmonary epithelial cells.

Cell culture and treatment - This experiment involved the human airway epithelial cell line, A549 (American Type Culture Collection, Bethesda, MD). The cells were cultured and treated as previously described in Example 2. Mite allergen was added to the culture medium to achieve a final concentration of 1000 ng/mL. Twenty-four hours later, the culture medium was sampled for PGE2 concentration.

PGE2 assay - Determination of PGE2 in the culture medium was performed as previously described in Example 1.

Statistical analysis - Means of eight replicates per treatment were computed using Excel spreadsheets (Microsoft, Redmond, WA).

Results Mite allergen treatment increased PGE2 biosynthesis 6-fold in A549 cells relative to the solvent treated controls (Figure 8).

HOPS DERIVATIVES INHIBIT MITE DUST ALLERGEN ACTIVATION OF

Summary - This example illustrates that hops derivatives are capable of inhibiting the PGE2 stimulatory effects of mite dust allergens in A549 pulmonary cells.

Methods The cell line and testing procedures are as described in Example 22. In addition to mite dust allergen, test materials included Hops fractions (1) alpha hop (1%
alpha acids; AA), (2) aromahop OE (10% beta acids and 2% isomerized alpha acids, (3) isohop (isomerized alpha acids; IAA), (4) beta acid solution (beta acids BA), (5) hexahop gold (hexahydro isomerized alpha acids; HHIAA), (6) redihop (reduced isomerized-alpha acids; RIAA), and (7) tetrahop (tetrahydro-iso-alpha acids THIAA).
Test materials at a final concentration of 10 g/mL were added 60 minutes prior to the addition of the mite dust allergen.

Results Table 15 depicts the extent of inhibition of PGE2 biosynthesis by hops derivatives in A549 pulmonary cells stimulated by mite dust allergen. All hops derivatives were capable of significantly inhibiting the stimulatory effects of mite dust allergens.

Table 15. PGE2 inhibition by hops derviatives in A549 pulmonary epithelial cells stimulated by mite dust allergen Test Material Percent Inhibition of PGE2 Bias' thesis Alpha hop AA 81 Aromahop OE 84 -1sohop (IAA) 78 Beta acids (BA) 83 Hexahop 82 Redihop 81 Tetrahop THIAA 76 In conclusion, it would also be useful to identify a natural formulation of compounds that would inhibit expression of COX-2, inhibit prostaglandin synthesis selectively in target cells, or inhibit inflammation response selectively in target cells.
A preferred embodiment comprises compositions containing at least one fraction isolated or derived from hops (Humulus lupulus). Examples of fractions isolated or derived from hops are alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.
Preferred compounds of fractions isolated or derived from hops, include, but are not limited to, humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. Preferred compounds can also bear substituents, such as halogens, ethers, and esters.

Another embodiment comprises composition containing tryptanthrin and conjugates thereof.

Other embodiments relate to combinations of components. One embodiment relates to compositions that include, as a first component, an active ingredient isolated or derived from an extract of hops and as a second component at least one member selected from the group consisting of rosemary (Rosmarinus officinalis L.), an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, and tryptanthrin or conjugates thereof. Another embodiment relates to compositions that include, as a first component, tryptanthrin or conjugates thereof and as a second component at least one member selected from the group consisting of an active ingredient isolated or derived from an extract of hops, rosemary, an extract or compound derived from rosemary, and a triterpene species or derivatives or conjugates thereof.

EFFECT OF MODIFIED HOPS COMPONENT ON NF-kB

As stated above, NF-KB, a heterodimer of the proteins p50 and Re1A, is an inducible eukaryotic DNA binding protein complex that is broadly expressed and plays a pivotal role in regulating multiple biological responses, such as the inflammatory and immune responses in mammalian cells. Targets of NF-KB include IL-2, the IL-2 receptor, and acute-phase proteins of the liver. In addition to its role in immune responses, NF-icB activation overrides the apoptotic response to TNF
and Fas, allowing for proliferation instead. NF-KB is cytoplasmic when inactive, maintained there by I-KB. As shown in Figure 9, various stimuli lead to activation of IKK (IxB Kinase), which phosphorylates bcB, marking it for ubiquitination and degradation. Once IKB is degraded, NF-xB is freed to initiate transcription.
Following transcriptional activation of a gene, NF-KB is also rapidly degraded.

The ability to detect activated or nondegraded NF-KB is a function of timing.
Following cytokine stimulation of a cell, activated NF-KB can be detected within 30 minutes. Within hours, the activated NF-KB is degraded and only nonactivated, complexed NF-KB remains. In this example, whole cell NF-kB was determined 24 hours following trivalent stimuli of Interluken-1f3 (IL-113), y-interferon (IFN), and TNFa. By this time, activated NF-KB has been degraded and only nonactivated, bound NF-KB remains. The IkB inhibitor is removed with lysis buffer and NF-KB
is quantified by enzyme immunoassay following capture by dsDNA containing the NF--KB response element. Compounds or mixtures that inhibit NF--KB activation can be identified as producing an increase in NF-xB-associated color development in cell lysates that have been treated with cytokines and the test material. Since NF-KB plays a key role in regulating both inflammatory and immune responses in mammalian cells, as well as contributing to cancer cell growth and increased replication of various viruses like HIV-1, the development of agents that impair NF-xB activation or function could have important therapeutic applications.

Methods Chemicals - NF-xB EIA kits were obtained from Active Motif (Carlsbad, CA).
Heat inactivated Fetal Bovine Serum (FBS-HI Cat. #3 5-011 CV), and Dulbeco's Modification of Eagle's Medium (DMEM Cat #10-013CV) was purchased from Mediatech (Herndon, VA). Reduced isomerized-alpha acids (RIAA) were obtained from John I. Haas, Inc., Yakima, WA. Interluken-1B (IL-1B), y-inteferon (IFN), TNFa, vitamin D3 (VD3) and all standard chemicals were obtained from Sigma (St Louis, MO) and were of the highest purity commercially available.

Cell culture and treatment of cells - The human monocytic cell line U937 was obtained from the American Type Culture Collection (Manasas, VA) and subcultured according to instructions from the supplier. The cells were routinely cultured at 37 C
with 5% CO2 in RPMI 1640 (Life Technologies, Grand Island, NY) containing 10%
FBS, with 50 units penicillin/mL, 50 g streptomycin/mL, 5% sodium pyruvate, and 5% L-glutamine (Life Technologies). For the experiment, U937 cells were cultured in 6-well plates at 37 C with 5% CO2 in a humidified incubator for 24 hours prior to treatment with test agents. RIAA in dimethylsulfoxide (10 L) was added to the cells to achieve a final concentration of 10 1g RIAA/mL 60 min prior to stimulation with VD3 (100 nM) or VD3 and the cytokine mixture (25 ng IL-IB, 150 ng IFN, and 20 ng TNFa/mL). Twenty-four hr later, the cells were washed and lysed with reagents supplied with the TransAM NFkB Chemi kit.

Protein assay - Protein concentrations of cell lysates were determined using the NanoOrange Protein Quantitation Kit with bovine serum albumin as the standard (Molecular Probes, Eugene, OE) according to the procedure supplied by the manufacturer. Fluorescence was determined using a Packard FluoroCount, Model BF
10000 fluorometer with the excitation filter set at 485 nm and emission filter set at 570 nm using Packard PlateReader version 3.0 software. The I-Smart program provided with the Packard PlateReader was used to calculate the protein concentration.

NF-,vB assay - The TransAM NFkB Chemi kit (Active Motiff) was used to detect nondegraded NF-icB p50 in the U937 cells. Instructions of the supplier were followed with no modification. A Bio-tek Instruments ELISA plate reader was used to record optical density at 405 nm. NF-xB was quantified and tabulated as mOD4os units.

Statistical analysis - Means of four to eight replicates per treatment and 95%
confidence intervals were computed using standard statistical formula in Excels spreadsheets (Microsoft, Redmond, WA).

Results After 24 hr of stimulation with the cytokine cocktail, the amount of nondegraded NF-icB in the U937 cells would be expected to decrease. As shown in Table 16, controls (330 mOD units) and VD3 treatments contained approximately twice the amount of NF--KB as those cells stimulated with the cytokine cocktail or VD3 plus cytokine cocktail. The addition of RIAA, however, prevented the activation and subsequent degradation of NF-KB (281 vs 122 mOD units). This was a novel and unexpected finding. The lack of activation of NF-xB is favorable because NF-KB
activation overrides the apoptotic response to TNF and Fas and can cause a variety of disorders. Also shown in Table 16, with RIAA alone, there was no activation of the NF-KB complex. Therefore, hops components or modified hops components, such as RIAA, can serve to affect disorders associated with NF-KB activation since these components either do not activate NF-KB or prevent activation of NF-KB.
Table 16 Treatment NF-KB
mOD units ]f Control (Dimethylsulfoxide solvent controls) 330 Vitamin D3 100 nM 391 261- 420 IL- 113/ -interferon/TNFa 25/150/20 n /mL 167 110 - 224 VD3 plus IL-113/ -interferon/TNFa 122 (79 - 165 Reduced isomerized alpha-acids 10 /mL 362 (301 422) RIAA/ VD3/IL-113/ -interferon/TNFa 281 (241 321) 'Parenthetic values are 95% confidence intervals.

In conclusion, it would be useful to identify a natural formulation of compounds that would to modulate NF-KB. Such a formulation has widespread applications.
It would also be useful to identify a natural formulation of compounds that would inhibit expression of COX-2, inhibit prostaglandin synthesis selectively in target cells, or inhibit inflammation response selectively in target cells.

ACIDS OR ISOMERIZED ALPHA ACIDS IN LPS-STIMULATED RAW 264.7 CELLS

This example describes testing of post COX-2 (cyclooxygenase-2) induction with A23187 arachidonic acid release.

The objective of this study was to assess the ability of the hops derivatives RIAA (reduced isomerized alpha acids) (Redihop (rho-iso-alpha acids (RIAA), 29.5 -30.5%, <0.2% iso-alpha acids) and IAA (isomerized alpha acids) (Isohop; iso-alpha acids (IAA), 29.5 - 30.5%) to function independently as direct inhibitors of mediated PGE2 biosynthesis in the RAW 264.7 cell model of inflammation.

The following methods and procedures were used. Cell culture and treatment with test material - RAW 264.7 cells (ATCC number TIB-71) were obtained from the American Type Culture Collection (Manassas, VA) and sub-cultured according to the instructions of the supplier. In preparation for testing, cells were grown in growth Dulbecco's Modification of Eagle's medium (DMEM) with 10% fetal bovine serum, heat inactivated (FBS-Hl) with penicillin/streptomycin and maintained in log phase prior to experimental setup. On day two of the experiment, cells were plated at 8 x 104 cells per well in a 96-well tissue culture plate with 200 L growth medium per well.

Following overnight incubation at 37 *C with 5% CO2, the growth medium was aspirated and replaced with 200 L DMEM with no FBS or penicillin/streptomycin.
RAW 264.7 cells were stimulated with lipopolysaccharide (LPS) (10 ng/ml final concentration) and incubated overnight to induce COX-2 expression. Eighteen hours post LPS-stimulation, test material was added followed 60 minutes later by the addition of A23187. Test materials were dissolved in dimethylsulfoxide (DMSO) as a 250-fold stock solution. Four L of this 250-fold stock test material preparation was added to 1 mL of DMEM, and 200 gL of this solution was added to eight wells for each dose of test material. Supernatant media was sampled for prostaglandin E2 (PGE2) determination after 30 minutes. Median inhibitory concentrations were computed from a minimum of four concentrations over two independent experiments.
The combination indeces (CIs) were computed as described below in statistical methods.

Table 17 describes the test materials used in each of two independent assays that were performed.

Table 17 Dosing matrix for LPS-stimulated RAW 264.7 cells followed by treatment with test material PGE2 assay using undiluted and 1:20 dilution - 2 plates 9.08 .03 -RAW 284.7 cells treated with LPS, incubated overnight then treated with test material for 60 min followed by A23187 for 30 min maound y~ctonRIAA [ug/inL] [lkgtmL] [IAglmt] Gg;mL] No, Wells 1. BetaTech RIAA 1.00 100 10 1.0 0.10 8 C7 CE c~a Ci C
I ej 'w I [Pg/ITII.J 1'm=j [u)'mL,' N
2. BetaTech IAA 1.00 100 10 1.0 0.10 8 C11 Aspirin 1.00 10 1.0 0.10 0.010 8 C12 APHS 10 1.0 0.10 0.010 8 Determination of PGE2 - A commercial, non-radioactive procedure for quantification of PGE2 was employed (Caymen Chemical, Ann Arbor, MI) for the determination of PGE2 and the recommended procedure of the manufacturer was used without modification. In summary, 50 L of the supernatant culture medium were diluted with appropriate amounts of acetylcholinesterase-labeled tracer and antiserum, and incubated at room temperature for 18 h. Afterwards, the wells in the PGE2-assay microtiter plate were emptied and rinsed with wash buffer; two-hundred gL of Ellman's reagent containing substrate for acetylcholinesterase were then added.
The reaction was maintained on a slow shaker at room temperature for 1 h and the absorbance at 415 nm was determined in a Bio-tek Instruments (Model #E1x800, Winooski, VT) enzyme-linked immunosorbant assay (ELISA) plate reader. The manufacturer's specifications for this assay include an intra-assay coefficient of variation of <10%, cross reactivity with PGD2 and PGF2 of less than 1% and linearity over the range of 10 - 1000 pg mL"1. The PGE2 concentration was computed as pg PGE2 per 105 cells.

Cell viability - Cell viability was assessed by microscopic inspection of cells prior to or immediately following sampling of the medium for PGE2 assay. Cell mortality was noted when observed.

The following materials were used and obtained by the indicated manufacturers. Bacterial lipopolysaccharide (LPS; B E. coli 055:B5) was from Sigma (St. Louis, MO). Prostaglandin E2 monoclonal antibody kit was purchased from Cayman Chemical (Ann Arbor, MI). Heat inactivated Fetal Bovine Serum (FBS-HI
Cat. #35-011CV) and Dulbecco's Modification of Eagle's Medium (DMEM Cat #10-1013CV) was purchased from Mediatech (Herndon, VA). Unless otherwise noted, all standard reagents were obtained from Sigma (St. Louis, MO) and were the purest commercially available. Test substances included RIAA and IAA obtained from Betatech Hops Products (Washington, DC).

The following statistical methods were used. A minimum of four concentrations (Table 1) was used to compute dose-response curves and medium inhibitory concentrations (IC5o5) for PGE2 with 95% confidence intervals using CalcuSyn (BIOSOFT, Ferguson, MO). This statistical package performs multiple drug dose-effect calculations using the Median Effect methods described by T-C
Chou and P. Talaly, Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 22, 27-55 (1984).
Briefly, the analysis correlates the "Dose" and the "Effect" in the simplest possible form: fa/fu = (C/Cm)m, where C is the concentration or dose of the compound and Cm is the median-effective dose signifying the potency. Cm is determined from the x-intercept of the median-effect plot. The fraction affected by the concentration of the test material is fa and the fraction unaffected by the concentration is fu (fu =1-fa). The exponent in is the parameter signifying the sigmoidicity or shape of the dose-effect curve. It is estimated by the slope of the median-effect plot.

The median-effect plot is a graph of x= log(C) vs. y = log(fa/fu) and is based on the logarithmic form of Chou's median-effect equation. The goodness of fit for the data to the median-effect equation is represented by the linear correlation coefficient r of the median-effect plot. Usually, the experimental data from enzyme or receptor systems have an r > 0.96, from tissue culture an r > 0.90 and from animal systems an r > 0.85. In the cell-based studies reported here, all linear correlation coefficients were greater than 0.90. For most robust results, experiments are repeated a minimum of three times on three different dates. The percent inhibition at each dose is averaged over the three independent experiments and used to calculate the median inhibitory concentrations reported.

Synergy of test components is quantified using the combination index (Cl) parameter. The CI of Chou-Talaly is based on the multiple drug-effect and is derived from enzyme kinetic models (Chou, T.-C. and Talalay, P. A simple generalized equation for the analysis of multiple inhibitions of Michaelis-Menten kinetic systems.
J. Biol. Chem. 252:6438-6442 (1977). The equation determines only the additive effect rather than synergism or antagonism. However, synergism is defined in this case as a more than expected additive effect, and antagonism as a less than expected additive effect as proposed by Cho and Talalay. Using the designation of Cl =
1 as the additive effect, the following relationships were obtained for mutually exclusive compounds that have the same mode of action or for mutually non-exclusive drugs that have totally independent modes of action: CI < 1, = 1, and > 1 indicating synergism, additivity and antagonism, respectively.

Two data transformations were applied where warranted. The first transformation consisted of computing the percent inhibition from the highest production produced from the lowest test concentration when the PGE2 production of these low doses exceeded the PGE2 production of the LPS-stimulated control.
This process controls for response variability and gradients throughout the plate.
The second data transformation adjusted for variance in response at the graded doses.
Monte Carlo simulations using the historical variance between wells predicted that dose-response curves appear graded only 40% of the time when duplicate wells per concentration are used in a four-point dose-response curve. Thus, sorting the response by concentration before calculating the IC50 was done in those situations in which the response did not appear graded.

Using the protocol outlined above, LPS-stimulation of PGE2 production in RAW 264.7 cells ranged from 1.4-fold to 2.1-fold relative to non-stimulated cells and was somewhat dependent upon dilution of media for the PGE2 assay. The IC50 value of 8.7 gg/mL (95% CL (confidence limit) = 3.9 - 19) computed for the aspirin positive control was consistent with published values for direct COX-2 inhibition ranging from 1.4 to 50 gg/mL (Mitchell, J.A. et al. Selectivity of nonsteroidal anti-inflammatory drugs as inhibitors of constitutive and inducible cyclooxygenase.
Proc.
Natl. Acad. Sci. USA 90:11693-11697 (1994); Warner, T.D. et al. Nonsteroidal drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: A full in vitro analysis. Proc. Natl. Acad.
Sci. USA
96:7563-7568 (1999)) and previous results of 3.2 g/mL (95% CL = 0.55 - 19) in the A549 cell line.

RAW 264.7 cells were stimulated with LPS and incubated overnight to induce COX-2 expression (Figure 10). Eighteen hours post LPS-stimulation, test material was added followed 60 minutes later by the addition of A23187. Supernatant media was sampled for PGE2 determination after 30 minutes. Mean percent PGE2 inhibition values were computed from a minimum of eight replicates over four concentrations and two independent experiments (Figure 10).

Both RIAA and IAA produced modest, dose-related inhibition of PGE2 in LPS-stimulated RAW 264.7 cells (Figure 10). Over the 1000-fold increase in concentration of test material, only a 14 and 10 percent increase in inhibition was noted, respectively, for RIAA and IAA. The shallowness of the dose-response slopes resulted in IC50 values (Table 18) in the mg/mL range for RIAA (36 mg/mL) and IAA

(>1000 mg/mL). Such minimal change in response over three-log units of doses implies that the observed inhibitory effect of the hops derivatives in this cell-based assay is likely a secondary effect on the cells and not direct inhibition of COX. One possible explanation is that the hops derivatives interfere with A23187-mediated arachidonic acid release from cellular membranes.

RAW 264.7 cells were stimulated with LPS and incubated overnight to induce COX-2 expression. Eighteen hours post LPS-stimulation, test material was added followed 60 minutes later by the addition of A23187. Supernatant media was sampled for PGE2 determination after 30 minutes. Median inhibitory concentrations were computed from a minimum of eight replicates at four concentrations over two independent experiments (Table 18).

Table 18. Median inhibitory concentrations for RIAA, IAA in RAW 264.7 cells when test material is added post overnight LPS-stimulation.

Test Material IC50 95% Confidence Interval m mL m /mL

IAA >1000 -Positive Control IC50 95% Confidence Interval ggAmLl /mL
Aspirin 8.7 gg/mL 3.9 - 19 The results of testing RIAA and IAA for their ability to directly inhibit PGE2 biosynthesis from COX-2 produced only a modest, dose-related inhibition of PGE2 in LPS-stimulated RAW 264.7 cells. The shallowness of the dose-response slopes resulted in IC50 values in the mg/mL range for RIA.A (36 mg/mL) and IAA (>1000 mg/mL). The observed inhibitory effect of the hops derivatives in this cell-based assay is likely a secondary effect on the cells and not direct inhibition of COX. One possible explanation is that the hops derivatives interfere with A23187-mediated arachidonic acid release from cellular membranes.

ANALYSIS OF HOPS ACTIVITY IN A CELL-FREE COX ASSAY SYSTEM
This example describes the effect of hops on COX enzyme activity.

The effect of hops on COX enzyme activity was tested in a cell-free COX
assay system. The assay was performed using the Cayman Chemical COX Inhibitor Screening Assay kit (Catalog # 560131; Cayman Chemical Co.; Ann Arbor MI).
Briefly, enzyme was pre-incubated with inhibitor for ten minutes at 37 C, then the reaction was initiated with the addition of arachidonic acid. After two minutes, the reaction was stopped by the addition of HCI. The PGH2 was reduced to PGA2 alpha, which was then quantified using a competitive enzyme immunoassay (EIA). Each concentration was tested with two separate reactions, which were then each tested in duplicate during the EIA step.

The results of the COX enzyme assay are shown in Table 19. As can be seen, IAA and RIAA had essentially no effect on COX-1 or COX-2 enzyme activity. In contrast, the COX inhibitor indomethacin inhibited both COX-1 and COX-2.

Table 19. IAA and RIAA inhibition of COX acitivity.

Indomethacin IAA RIAA

ug/mI % Inhibition ug/ml % Inhibition ug/ml % Inhibition 10 60.6 200 9.6 200 2.9 1 34.6 100 3 100 0.5 0.01 28.2 10 4.2 10 2.5 0.001 -2.8 1 2.1 1 1.9 ug/mi % Inhibition ug/ml % Inhibition ug/ml % Inhibition 200 48.4 200 0.3 200 1.9 50 77.6 100 -3.6 100 -0.7 0.5 67.5 10 -2 10 2.7 0.05 3.4 1 0.9 1 2.8 These results demonstrate that hops have no significant COX enzyme activity.
Therefore, hops likely acts at the level of expression of COX-2.

GASTRIC MUCOSAL CELL MODEL FOR ESTIMATING RELATIVE
GASTROINTESTINAL TOXICITY OF NSAIDS
This example describes the use of a gastric mucosal cell line (AGS cells) for determining potential gastrointestinal toxicity of non-steroidal anti-inflammatory drugs.

As described in Examples 1 and 2, AGS cells provide a model system for determining potential gastrointestinal toxicity. The objective of this study was to further characterize the AGS human gastric mucosal cell line as a model for estimating relative GI toxicity (gastropathy) of COX-inhibiting compounds.

AGS cells were further characterized as a model for gastrointestinal toxicity essentially as described in Example 2. Briefly, chemicals used in the assays were obtained as follows. Commercial formulations of rofecoxib tablets and celecoxib capsules were used. PGE2 EIA kits were obtained from Cayman Chemical (Ann Arbor, MI). Anti-COX-1 and anti-COX-2 rabbit polyclonal antisera were obtained from Upstate Biotechnology (Waltham, MA), and donkey anti-goat IgG-HRP was procured from Santa Cruz Biotechnology (Santa Cruz, CA). Heat Inactivated Fetal Bovine Serum (FBS-HI Cat. #35-01 1CV) and Dulbecco's Modification of Eagle's Medium (DMEM Cat #10-013CV) was purchased from Mediatech (Herndon, VA).
Interleukin-1(3 (IL-113) and all standard chemicals and non-steroidal anti-inflammatory drugs (NSAIDs), unless noted, were obtained from Sigma (St Louis, MO) and were of the highest purity commercially available.

For cell culture, human airway epithelial cells A549 were obtained from the American Type Culture Collection (Manassas, VA) and sub-cultured according to the instructions of the supplier. The cells were routinely cultured at 37 C with 5% CO2 in RPMI 1640 containing 10% FBS with 50 units penicillin/mL, 50 g streptomycin/mL, 5% sodium pyruvate, and 5% L-glutamine. For treatment of the cells with test compounds, the William Harvey Modified Assay (WilMA) was used for' determination of COX-2 inhibition with no modifications [Warner TD, Giuliano F, Vojnovic I, Bukasa A, Mitchell JA, Vane JR. (1999) Non-steroidal drug selectivities for cyclooxygenase-1 rather than cyclooxygensase-2 are associated with human gastrointestinal toxicity: A full in vitro analysis. Proc. Natl. Acad. Sci.
USA.
96:7563-7568]. Briefly, A549 cells were exposed to IL-113 for 24 h, media was removed and human plasma (100 L) was added together with test agents or the DMSO vehicle. For initial experiments, test agent concentrations were 25, 5, 0.5, and 0.05 g/mL. DMSO in each microtiter well was less than 1% of the 200 L
volume;
60 min later, A23187 (50 M) was added; after 30 min, 50 L of the culture supernatant media was sampled and immediately assayed for PGE2 as detailed elsewhere. The AGS human gastric mucosal cell line (American Type Culture Collection, Manassas, VA) was also cultured and maintained according to recommended ATCC methodology. Sub-cultured AGS cells were grown in IMDM
with 20% FBS with 50 units penicillin/mL and 50 gg streptomycin/mL; cells were maintained in log phase prior to each experiment. For PGE2 assays, approximately 105 cells per well were plated into 96-well plates in 200 L growth medium per well.
Cells were grown to 80% confluence and washed 3-times with IMDA media prior to addition of test agent. NSAIDs were added in 200 L of IMDA media containing no FBS or penicillin/streptomycin. Sixty minutes following addition of the test materials, arachidonic acid (AA) was either induced with addition of the calcium ionophore A23187, or exogenously added at 100 or 5 gM AA in DMSO. Incubation at 37 C
was carried out for an additional 30 min. Fifty microliters of media were sampled for PGE2 determination.

For determination of PGE2, a commercial, non-radioactive procedure for quantification of PGE2 was employed (Cayman Chemical, Ann Arbor, MI) for the determination of PGE2 and the recommended procedure of the manufacturer was used without modification. Briefly, 50 L of the supernatant culture medium, along with a serial dilution of PGE2 standard samples, were mixed with appropriate amounts of acetylcholinesterase-labeled tracer and PGE2 antiserum and incubated at room temperature for 18 h. Afterwards, the wells in the PGE2-assay microtiter plate were emptied and rinsed with wash buffer, and 200 L of Ellman's reagent containing substrate for acetylcholinesterase were then added. The reaction was performed on a slow shaker at room temperature for 1 h, after which absorbance at 415 nm was determined in a Bio-Tek Instrument ELISA plate reader (Model #Elx800, Winooski, VT). The manufacturer's specifications for this assay include an intra-assay coefficient of variation of < 10%, cross reactivity with PGD2 and PGF2a, of less than 1%, and linearity over the range of 10-1000 pg/mL. The PGE2 concentration was recorded as pg PGE2 per 105 cells.

For assay calculations, a minimum of four concentrations each, with two replicates per concentration over three independent experiments, were used to compute median inhibitory concentrations (IC50) and their 95% confidence limits for the inhibition of PGE2 biosynthesis (CalcuSyn, BIOSOFT, Ferguson, MO).
Complete dose-response curves with coefficients of determination > 0.9 were obtained for all test compounds, except salicylic acid and acetaminophen for A549 and AGS cells and diisopropylfluorophosphate for AGS cells with 100 M AA. The therapeutic index (TI) for gastrointestinal safety was computed as the log (AGS IC50/A549 IC50).
While it has been recommended that the IC80 should be used for the efficacy component (i.e., inhibition of PGE2 synthesis in A549 cells) in the calculation of the TI, large errors are associated with estimates at the extremes of the dose-response curve. Thus, there exists greater uncertainty in ratios in which these estimates are used.
Positive TI
indicate low potential for GI toxicity, while negative TI indicate a higher potential for GI toxicity. Spearman's rank correlation coefficient rs was computed to quantify the degree of association between ranking of previously published TI using non-target cells and the AGS/A549 model. The parameter rs was also used to determine the degree of association between in vitro ranking of TI and ranking of clinically assessed NSAID gastropathy. The probability of a Type I error was set at the nominal 5%
level.

The AGS human gastric cell line was cultured in six-well plates at 37 C with 5% C02 in a humidified incubator for 24 h. Cells were lysed on ice in lysis buffer and protein concentration determined. Fifty micrograms of cell lysate were solubilized and fractionated on a 10% polyacrylamide gel containing sodium dodecylsulfate. For immunoblotting, Western blotting of COX-1 and COX-2 was performed using PAGErTM Gold Precast Gels (Bio Whittaker Molecular Applications, Rockland, ME). RAW 264.7 cell lysates containing approximately 60 gg protein were loaded with laemmli sample buffer into the wells in a total volume of 30 L.
The vertical minigel electrophoresis chambers used were made by Savant Instruments, Inc. (Model MV 120; Holbrook, NY). Gels were run at 40 mA/plate (constant current) at room temperature until the bromophenol blue stain reached the bottom of the gel, which took about 1 h. Gels were then blotted on polyvinyl fluoride transfer membranes (PVDF) (Pall Corporation, Ann Arbor, MI), overnight, at 500 mA and 4 C. Molecular weight markers used were the unstained, broad-range Precision Protein Standards (Bio Rad, Hercules, CA). The BioWestTM extended duration chemiluminescent substrate (Biolmaging Systems, Upland, CA), a non-isotopic, horseradish peroxidase substrate kit for Western Blot detection, was used for protein visualization. Images of Western Blots were acquired using a UVP Epi Chemi II
Darkroom (Biolmaging Systems), and were analyzed and enhanced by LabWorksTM
Image Acquisition and Analysis Software (Biolmaging Systems). Band intensities were evaluated through densitometric analysis, computed using ScanAnalysis software (BIOSOFT, Ferguson, MO), and recorded as arbitrary Density Units (DU).
The AGS cell line constitutively expressed both COX-1 and COX-2, with COX-1 expression approximately 4 times greater than COX-2 expression (see Example 1). These results are in agreement with recently published work (Fan et al., Interleukin- I beta induces cyclo-oxygenise-2 expression in gastric cancer cells by the p38 and p44/42 mitogen-activated protein kinase signaling pathways. J
Gastroenterol Hepatol. 16:1098-104 (2001)) using the AGS cell line, in which constitutive expression of both cyclooxygenase enzymes was demonstrated.

Table 20 shows the median inhibitory concentration (IC50) for PGE2 biosynthesis of select NSAID in the A549 and AGS cell model.

Table 20. Median inhibitory concentrations (IC50) for PGE2 biosynthesis of select NSAID in the A549 and AGS cell model.t Compounds A549 AGS Gastric Mucosal Cells [11M] [11M] Arachidonate Arachidonate DIFPtt 6.5 359 >136 217 (1.5-28) 125-1022 (141-185) Rofecoxib 0.24 5.5 12 21 (0.15-0.45) (2.7-11) (2.3-64) (5.8-79) Celecoxib 0.21 0.063 17 9.4 0.01-4.2 (0.02-0.22) 2.5-113 (3.9-23) Nimensulide 0.32 0.12 73 75 (0.16-0.65) (0.0081-1.7) (25-211) (52-104) Naproxen 28 0.83 167 313 (1.3-600) (0.24-2.8) (16-1735) (91-1039) Ibuprofen 12 2.8 6.3 107 (6.8-19) (1.3-5.8) (1.4-29) (38-291) Aspirin 18 2.9 6.0 18 (3.0-106) 1.4-5.6 (2.9-12) (9.4-37) Salicylic acid 4246 1065 112 7848 (355-50971) (94-12217) (69-181) (1775-34565) Acetaminophen 238 535 346 3815 (6.62-9589) (179-1616) (192-609) (1152-12649) Indomethacin 8.1 0.0042 0.0025 0.0056 (2.6-26) (0.00042-0.039) (0.000028-0.20) (0.002-0.011) t Parenthetic values are 95% confidence intervals of the IC50 estimate.
tt DIFP =.diisofluorophosphate.

Figure 11 shows a comparison of Log IC50 ratios and ranking of potential gastropathy. Log IC50 ratios using the William Harvey Modified Assay (WHMA) are expressed WHMA COX-1/WHMA COX-2 from Warner, et al. (Nonsteroidal drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygensase-2 are associated with human gastrointestinal toxicity: A full in vitro analysis. Proc. Natl. Acad.
Sci. USA.
96:7563-7568(1999))(white bars) and Mitchell, et al. (Selectivity of nonsteroidal antiinflammatory drugs as inhibitors of constitutive and inducible cyclooxygenase.
Proc. Natl. Acad. Sci. USA. 90:11693-11697 (1994))(blue bars) are shown in Figure 1 1A, with log IC50 ratios (AGS/WHMA COX-2) for AGS cells treated with A23187 Figure 1 1B), 100 M arachidonic acid (Figure 11C), or 5 M arachidonic acid (Figure 11D). Values to the right of 0 indicate decreasing probability of gastropathy, whereas values to the left of 0 indicate increasing probability of gastropathy.

As shown in Figure 11, the relationship between previously published data and the three AGS protocols was examined. The log[IC5o ratio(WHMA COX-1/WIIMA
COX-2)] was computed from published data (Figure 11A) and compared to the log[IC5o ratios(AGS/WIINIA COX-2)] for the A23187 (Figure 11B), 100 M
arachidonic acid (Figure 11C), and 5 M arachidonic acid (Figure 11D) protocols.
Ranking of compounds from lowest to greatest GI toxicity potential was strikingly similar among the models and within AGS protocols. Interestingly, only rofecoxib and indomethacin showed the least and most potential GI toxicity, respectively, in all AGS protocols. Rank estimates of TI were similar to rank TI computed using platelets (COX-1) and A549 cells (COX-2) with rs 0.903, p<0.01 for A23187;
0.733, p=0.02 for 100 M AA; and 0.77, p<0.02 for 5 gM AA. Quantitatively, however, the AGS/A549 model exhibited lower TI than the platelet/A549 model. A23187-mediated AA release, 100 gM AA addition, and 5 M AA addition provided rank estimates of GI toxicity significantly associated with clinical rankings of NSAID
gastropathy, respectively (rs 0.933, p<0.01; 0.783, p<0.01; 0.683, p=0.05).

ranking of NSAIDs from lowest to greatest potential for GI toxicity was rofecoxib <
acetaminophen < nimensulide < celecoxib < salicylic acid < ibuprofen < aspirin <
naproxen < indomethacin.

As described in Example 1 and as seen in normal gastric mucosal cells, the AGS cell line constitutively expresses both COX-1 and COX-2. The use of AGS
and A549 cells provided rank estimates of TI that were qualitatively similar to rank TI
computed using platelets (COX-1) and A549 cells with rs = 0.903, p<0.01 for A23187; 0.733, p=0.02 for 100 M AA; and 0.770, p<0.02 for 5 gM AA.
Quantitatively, the AGS/A549 model exhibited lower TI than the platelet/A549 model. A23187-mediated AA release, as well as the 100 gM and 5 gM AA-addition protocols provided rank estimates of GI toxicity significantly associated with clinical rankings of NSAID gastropathy, at r,=0.933, p<0.01; 0.783, p<0.01; and 0.683, p=0.05, respectively. A23187 ranking of NSAIDs from lowest to greatest potential for GI toxicity was rofecoxib < acetaminophen < nimensulide <celecoxib <
salicylic acid < ibuprofen < aspirin < naproxen < indomethacin.

These results further demonstrate that the AGS human gastric mucosal cell line can serve as a model for assessing the gastrointestinal effects of COX-inhibiting compounds. The AGS derived data indicate that inhibition of gastric PGE2 biosynthesis underlies the human gastropathy of NSAIDs.

A preferred embodiment comprises compositions containing at least one fraction isolated or, derived from hops (Humulus lupulus). Examples of fractions isolated or derived from hops are alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops.
Preferred compounds of fractions isolated or derived from hops, include, but are not limited to, humulone, cohumulone, adhumulone, isohumulone, isocohumulone, isoadhumulone, dihydro-isohumulone, dihydro-isocohumulone, dihydro-adhumulone, tetrahydro-isohumulone, tetrahydro-isocohumulone, tetrahydro-adhumulone, hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone. Preferred compounds can also bear substituents, such as halogens, ethers, and esters.

Another embodiment comprises composition containing tryptanthrin and conjugates thereof.

Other embodiments relate to combinations of components. One embodiment relates to compositions that include, as a first component, an active ingredient isolated or derived from an extract of hops and as a second component at least one member selected from the group consisting of rosemary (Rosmarinus officinalis L.), an extract or compound derived from rosemary, a triterpene species or derivatives or conjugates thereof, and tryptanthrin or conjugates thereof. Another embodiment relates to compositions that include, as a first component, tryptanthrin or conjugates thereof and as a second component at least one member selected from the group consisting of an active ingredient isolated or derived from an extract of hops, rosemary, an extract or compound derived from rosemary, and a triterpene species or derivatives or conjugates thereof.

It will be readily apparent to those skilled in the art that various changes and modifications of an obvious nature maybe made without departing from the spirit of the invention, and all such changes and modifications are considered to fall within the scope of the invention as defined by the appended claims. Such changes and modifications would include, but not be limited to, the incipient ingredients added to affect the capsule, tablet, lotion, food or bar manufacturing process as well as vitamins, herbs, flavorings and carriers. Other such changes or modifications would include the use of other herbs or botanical products containing the combinations of the preferred embodiments disclosed above. Many additional modifications and variations of the embodiments described herein may be made without departing from the scope, as is apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only.

Claims (14)

The embodiments of the present invention for which an exclusive property or privilege is claimed are defined as follows:
1. A composition comprising as a first component, 0.5 to 10000 mg of a fraction isolated or derived from hops; and as a second component, at least one member selected from the group consisting of 0.5 to 5000 mg of a compound derived from rosemary, and 0.035 to 3500 mg of a triterpene species; wherein a. the fraction isolated or derived from hops is selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops;

b. the compound derived from rosemary is selected from the group consisting of carnosic acid, oleanolic acid, rosemarinic acid, and ursolic acid; and c. the triterpene species is selected from the group consisting of oleanolic acid and ursolic acid.
2. The composition of claim 1, wherein the fraction isolated or derived from hops is extracted with CO2.
3. The composition of claim 1 or claim 2, wherein the composition comprises about 0.001 to 10 weight percent of the first component.
4. The composition of any one of claims 1 to 3, wherein the composition comprises about 0.001 to 10 weight percent of the second component.
5. The composition of claim 1 or claim 2, wherein a ratio of the first component to the second component is in the range of about 100:1 to about 1:100.
6. The composition of any one of claims 1 to 5, wherein the composition further comprises a pharmaceutically acceptable carrier.
7. The composition of any one of claims 1 to 6, further comprising glucosamine.
8. Use of a composition for inhibiting an inflammatory response in cells, wherein said composition comprises as a first component a fraction isolated or derived from hops and a second component selected from the group consisting of a compound derived from rosemary, and a triterpene species; wherein a. the fraction isolated or derived from hops is selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops;

b. the compound derived from rosemary is selected from the group consisting of carnosic acid, oleanolic acid, rosemarinic acid and ursolic acid; and c. the triterpene species is selected from the group consisting of oleanolic acid and ursolic acid.
9. The use of claim 8, wherein the composition further comprises glucosamine.
10. Use of a composition for treating or inhibiting a pathological condition in a mammal associated with tissue-specific activation of inflammation, wherein said composition comprises as a first component a fraction isolated or derived from hops and a second component selected from the group consisting of a compound derived from rosemary, and a triterpene species; wherein a. the fraction isolated or derived from hops is selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops;

b. the compound derived from rosemary is selected from the group consisting of carnosic acid, oleanolic acid, rosemarinic acid and ursolic acid; and c. the triterpene species is selected from the group consisting of oleanolic acid and ursolic acid.
11. The use of claim 10, wherein the composition comprises about 0.5 to 10000 mg of the fraction isolated or derived from hops.
12. The use of claim 10, wherein the composition comprises about 0.001 to 10 weight percent of the fraction isolated or derived from hops.
13. The use of any one of claims 10 to 12, wherein the composition further comprises a third component different from the second component, said third component is selected from the group consisting of a compound derived from rosemary, and a triterpene species.
14. Use of a composition for the manufacture of a medicament for inhibiting an inflammatory response in cells, wherein said composition comprises as a first component a fraction isolated or derived from hops and a second component selected from the group consisting of a compound derived from rosemary, and a triterpene species;
wherein a. the fraction isolated or derived from hops is selected from the group consisting of alpha acids, isoalpha acids, reduced isoalpha acids, tetra-hydroisoalpha acids, hexa-hydroisoalpha acids, beta acids, and spent hops;

b. the compound derived from rosemary is selected from the group consisting of carnosic acid, oleanolic acid, rosemarinic acid and ursolic acid; and c. the triterpene species is selected from the group consisting of oleanolic acid and ursolic acid.
CA2503196A 2002-10-21 2003-10-20 Compositions that treat or inhibit pathological conditions associated with inflammatory response Expired - Lifetime CA2503196C (en)

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
US42038302P 2002-10-21 2002-10-21
US60/420,383 2002-10-21
US45023703P 2003-02-25 2003-02-25
US60/450,237 2003-02-25
US40029303A 2003-03-26 2003-03-26
US40128303A 2003-03-26 2003-03-26
US10/400,293 2003-03-26
US10/401,283 2003-03-26
US10/464,834 US20040115290A1 (en) 2001-06-20 2003-06-18 Modulation of inflammation by hops fractions and derivatives
US10/464,410 US8142819B2 (en) 2002-10-21 2003-06-18 Synergistic compositions that treat or inhibit pathological conditions associated with inflammatory response
US10/464,410 2003-06-18
US10/464,834 2003-06-18
PCT/US2003/033362 WO2004037180A2 (en) 2002-10-21 2003-10-20 Compositions that treat or inhibit pathological conditions associated with inflammatory response

Publications (2)

Publication Number Publication Date
CA2503196A1 CA2503196A1 (en) 2004-05-06
CA2503196C true CA2503196C (en) 2011-08-02

Family

ID=32180835

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2503196A Expired - Lifetime CA2503196C (en) 2002-10-21 2003-10-20 Compositions that treat or inhibit pathological conditions associated with inflammatory response

Country Status (9)

Country Link
US (1) US8486457B2 (en)
EP (1) EP1558271A4 (en)
JP (1) JP2006508182A (en)
KR (1) KR20050071605A (en)
AU (1) AU2003286549B2 (en)
CA (1) CA2503196C (en)
MX (1) MXPA05004288A (en)
NZ (1) NZ539642A (en)
WO (1) WO2004037180A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110141572A (en) * 2019-06-20 2019-08-20 牡丹江医学院 A kind of drug and preparation method thereof for treating asthma

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7901714B2 (en) * 2001-06-20 2011-03-08 Metaproteomics, Llp Treatment modalities for autoimmune diseases
US7815944B2 (en) * 2001-06-20 2010-10-19 Metaproteomics, Llc Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment of prevention of gastric toxicity
US8168234B2 (en) 2001-06-20 2012-05-01 Metaproteomics, Llc Compositions that treat or inhibit pathological conditions associated with inflammatory response
US8206753B2 (en) * 2001-06-20 2012-06-26 Metaproteomics, Llc Anti-inflammatory botanical products for the treatment of metabolic syndrome and diabetes
US7901713B2 (en) * 2001-06-20 2011-03-08 Metaproteomics, Llc Inhibition of COX-2 and/or 5-LOX activity by fractions isolated or derived from hops
US8142819B2 (en) 2002-10-21 2012-03-27 Metaproteomics, Llc Synergistic compositions that treat or inhibit pathological conditions associated with inflammatory response
US20040115290A1 (en) * 2001-06-20 2004-06-17 Tripp Matthew L. Modulation of inflammation by hops fractions and derivatives
US8158160B2 (en) 2001-11-13 2012-04-17 Eric Hauser Kuhrts Anti-inflammatory cyclooxygenase inhibitors
NZ539642A (en) 2002-10-21 2007-01-26 Metaproteomics Llc Compositions containing extracts from hops in combination with a second component for treating inflammatory response
KR20060105429A (en) 2003-05-22 2006-10-11 메타프로테오믹스, 엘엘씨 Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity
US20050192356A1 (en) * 2004-02-27 2005-09-01 Babish John G. Synergistic anti-inflammatory pharmaceutical compositions and methods of use
US7914831B2 (en) * 2004-02-27 2011-03-29 Metaproteomics, Llc Synergistic anti-inflammatory pharmaceutical compositions and related methods using curcuminoids or methylxanthines
JP4321769B2 (en) * 2004-04-13 2009-08-26 Towa Corporation 株式会社 Composition for antihistamine, composition for prevention and treatment of allergic rhinitis, composition for prevention and treatment of hay fever and composition for treatment of urticaria
EP1817941A4 (en) * 2004-11-13 2009-11-11 Metaproteomics Llc Compositions exhibiting inhibition of cyclooxygenase-2
AU2006280074A1 (en) * 2005-08-09 2007-02-22 Metaproteomics, Llc Protein kinase modulation by hops and acacia products
AU2012204133B2 (en) * 2005-08-09 2013-09-19 Metaproteomics, Llc Protein kinase modulation by hops and acacia products
JP2009518429A (en) * 2005-12-09 2009-05-07 メタプロテオミクス,エルエルシー Anti-inflammatory plant products for the treatment of metabolic syndrome and diabetes
JP2009518439A (en) * 2005-12-09 2009-05-07 メタプロテオミクス,エルエルシー Protein kinase regulation by hops and acacia products
JP5066706B2 (en) * 2006-02-28 2012-11-07 国立大学法人徳島大学 Screening method for anti-obesity agents
EP2046353A4 (en) * 2006-06-20 2010-01-27 Metaproteomics Llc Tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
US20080051466A1 (en) * 2006-06-20 2008-02-28 Metaproteomics, Llc Isoalpha acid based protein kinase modulation cancer treatment
WO2008026473A1 (en) 2006-08-31 2008-03-06 Asahi Breweries, Ltd. Method for production of hop preparation, hop preparation, antiinflammatory agent, food/beverage, and oral product
KR20080025900A (en) * 2006-09-19 2008-03-24 (주) 서울바이오메드 A composition and a method for treatment of conjunctivitis comprising glucosamine and derivatives thereof
KR100893162B1 (en) * 2007-02-21 2009-04-17 바이오스펙트럼 주식회사 Agents for Improving Skin Wrinkles Comprising Carvacrol as an Active Ingredient
MX2009010049A (en) * 2007-03-19 2010-03-04 Metaproteomics Llc Methods and compositions for promoting bone and joint health.
WO2008140842A1 (en) * 2007-05-11 2008-11-20 Metaproteomics, Llc Methods and compositions for heavy metal detoxification
AU2008335156A1 (en) * 2007-12-10 2009-06-18 Metaproteomics, Llc Substituted 1,3-cyclopentadione multi-target protein kinase modulators of cancer, angiogenesis and the inflammatory pathways associated therewith
JP2011516492A (en) * 2008-04-02 2011-05-26 メタプロテオミクス, エルエルシー Attenuation of endothelial inflammation and endothelium-monocyte interaction by substituted 1,3-cyclopentadione
KR100919898B1 (en) * 2008-07-24 2009-09-30 바이오스펙트럼 주식회사 Anti-obesity Agents Comprising Carvacrol as an Active Ingredient
JP2010138155A (en) * 2008-12-15 2010-06-24 Pola Chem Ind Inc External preparation for skin suitable for ameliorating drabness
KR101050429B1 (en) * 2009-03-18 2011-07-19 연세대학교 산학협력단 Composition for the prevention or treatment of obesity, dyslipidemia, fatty liver or insulin resistance syndrome comprising camphor as an active ingredient
KR101148623B1 (en) * 2010-03-25 2012-05-21 대구한의대학교산학협력단 A COMPOSITION COMPRISING &alpha;-THUJONE FOR PREVENTING AND TREATING COGNITIVE DYSFUNCTION AND IMPROVING MEMORY
KR20140027910A (en) 2010-10-30 2014-03-07 킨덱스 테라퓨틱스, 엘엘씨 Cis 3,4-dihydroxy-2-(3-methylbutanoyl)-5-(3-methylbutyl)-4-(4-methylpentanoyl)cyclopent-2-en-1-one derivatives, substantially enantiomerically pure compositions and methods
JP2013184942A (en) * 2012-03-09 2013-09-19 Kose Corp Antioxidant and dna damage inhibitor
KR101936688B1 (en) 2012-04-17 2019-04-09 (주)아모레퍼시픽 Whitening composition containing trolox
DK2900230T3 (en) 2012-09-27 2018-11-12 Childrens Medical Ct Corp RELATIONSHIPS FOR TREATING ADIPOSITAS AND PROCEDURES FOR USING THEREOF
JP6163084B2 (en) * 2013-11-11 2017-07-12 キリン株式会社 Composition for preventing, treating and / or improving cognitive function of dementia, and medicine and food using the same
JP2017513824A (en) * 2014-03-26 2017-06-01 ザ チルドレンズ メディカル センター コーポレイション Celastrol and derivatives for the treatment of obesity
RU2549475C1 (en) * 2014-05-12 2015-04-27 Федеральное государственное бюджетное учреждение науки Тихоокеанский институт биоорганической химии им. Г.Б. Елякова Дальневосточного отделения Российской академии наук (ТИБОХ ДВО РАН) Pharmaceutical composition possessing therapeutic action on various skin pathologies
JP6242746B2 (en) * 2014-05-26 2017-12-06 キリン株式会社 Medicines, foods, and compositions that improve memory learning function
US9125912B1 (en) * 2015-02-10 2015-09-08 King Saud University Method of treating ulcerative colitis
KR102043893B1 (en) * 2018-01-05 2019-11-12 인제대학교 산학협력단 Composition Comprising alpha-Humulene for Preventing or Treating Diseases Associated with Mucous Membrane of Digestive System
JP2020028263A (en) * 2018-08-23 2020-02-27 国立大学法人徳島大学 Disuse muscular atrophy suppressing agent and food composition for disuse muscular atrophy suppression
CN112043713B (en) * 2020-09-14 2021-10-12 南通大学 Application of betulinic acid derivative in preparing medicine for treating nerve injury diseases
CN112625042B (en) * 2020-12-28 2022-04-01 北京燕化集联光电技术有限公司 Organic electroluminescent material and application thereof
CN112794872B (en) * 2021-04-02 2022-07-15 湖南德诺贝莱健康产业有限公司 Method for extracting luteolin-3' -glucuronide from rosemary
CN113350360A (en) * 2021-06-18 2021-09-07 西南医科大学 Novel application of triterpenic acid compound as A beta fiber formation inhibitor drug

Family Cites Families (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1901277U (en) 1964-05-25 1964-09-24 Kuhlmann Geb DEVICE FOR MAKING PERFORMANCE IN PROFILE BARS.
US3933919A (en) 1964-12-15 1976-01-20 Geoffrey Wilkinson Hydroformylation of mono-α-olefins and mono-α-acetylenes
US3451821A (en) 1965-03-01 1969-06-24 Kalamazoo Spice Extract Co Increasing the utilization of hops and improving flavor control of malt beverages and the like
GB1145240A (en) 1965-03-01 1969-03-12 Kalamazoo Spice Extract Co Hop flavours for malt beverages and the like
US3720517A (en) 1970-12-21 1973-03-13 Hamm T Brewing Co Preparation of a fermented malt champagne
US3932603A (en) 1971-05-28 1976-01-13 General Foods Corporation Oral preparations for reducing the incidence of dental caries
GB1372932A (en) 1971-03-15 1974-11-06 Gen Foods Corp Anti-caries compositions
US3965188A (en) 1972-01-10 1976-06-22 Miller Brewing Company Hop extract process and product
CH617326A5 (en) 1975-12-04 1980-05-30 Siegfried Ag
US4170638A (en) 1976-11-05 1979-10-09 S. S. Steiner, Inc. Method for producing a deodorant
US4148873A (en) 1976-11-05 1979-04-10 S. S. Steiner, Inc. Method for treating the skin with extracts of hops
US4123561A (en) 1977-02-01 1978-10-31 S.S. Steiner, Inc. Method for processing hops for brewing
US4401684A (en) 1981-10-01 1983-08-30 Australian Hop Marketers Pty. Ltd. Preservation of hops utilizing ascorbic acid
US4389421A (en) 1981-10-30 1983-06-21 Busch Industrial Products Corporation Method for controlling light stability in malt beverages and product thereof
BE896610A (en) 1982-05-06 1983-08-16 Hop Developments Ltd EXTRACTION OF PLANT MATERIAL USING CARBONIC ANHYDRIDE
US4486436A (en) 1982-07-22 1984-12-04 Analgesic Associates Analgesic and anti-inflammatory compositions comprising caffeine and methods of using same
US4473551A (en) 1982-08-23 1984-09-25 Faxon Pharmaceuticals, Inc. Anti-inflammatory composition
US4644084A (en) 1984-01-25 1987-02-17 Miller Brewing Company Preparation of tetrahydroisohumulones
GB2187755B (en) 1984-02-28 1990-03-28 Kalamazoo Holdings Inc Separation of the constituents of co2 hop extracts
SU1247011A1 (en) 1985-02-20 1986-07-30 Специальное Конструкторское Бюро Химизации Научно-Производственного Объединения "Аэрозоль" Agent for hair care
DE3513169A1 (en) 1985-04-12 1986-10-16 Hopstabil Hopfenverarbeitungs-Gesellschaft mbH, 8069 Wolnzach METHOD FOR PRODUCING ISOHUMULONES
US4767640A (en) 1985-10-29 1988-08-30 Miller Brewing Company Light stable hop extracts and method of preparation
US4692280A (en) 1986-12-01 1987-09-08 The United States Of America As Represented By The Secretary Of Commerce Purification of fish oils
US5041300A (en) 1987-04-03 1991-08-20 Kalamazoo Holdings, Inc. Hop flavor which is odor forming impurity free
DE3712986A1 (en) 1987-04-16 1988-10-27 Marbert Gmbh MEDICAL PREPARATIONS BASED ON TREASURE EXTRACT, METHOD FOR THE PRODUCTION THEREOF AND USE OF TREATMENT EXTRACT FOR THE PRODUCTION OF COSMETIC PREPARATIONS AND A SPECIAL TREATMENT EXTRACT
US4857554A (en) 1987-08-17 1989-08-15 Georgios Kallimanis Method for the treatment of psoriasis
US5082975A (en) 1988-08-15 1992-01-21 Kalamazoo Holdings, Inc. Synthesis of hexahydrolupulone, novel forms thereof, and its use as a selective inhibitor of cell growth and multiplication
US5166449A (en) 1988-08-15 1992-11-24 Kalamazoo Holdings, Inc. Synthesis of hexahydrolupulone, novel forms thereof, and its use as a selective inhibitor of cell growth and multiplication
DE3931147A1 (en) 1989-09-19 1991-03-28 Solong Natural Ltd New nerve tonic contg. extract of avena sativa - used to treat frigidity and increase libido in women
US5013571A (en) 1990-01-31 1991-05-07 Pfizer Inc. Methods for making tetrahydroisoalpha and hexahydroisoalpha acids
DE59010282D1 (en) 1990-09-10 1996-05-15 Fromm Mayer Bass Ltd A process for isomerizing humulon in a carbon dioxide hop extract and a process for obtaining isohumulone therefrom
JP3155003B2 (en) 1990-11-06 2001-04-09 サントリー株式会社 Method for producing hop extract and hop extract obtained by the method
TW199905B (en) 1992-02-03 1993-02-11 J E Siebel Sons Company Inc Method and composition for enhancing foam properties of fermented malt beverages
RU2045955C1 (en) 1992-02-24 1995-10-20 Никитина Татьяна Ивановна Method for treating adnexites
US5641517A (en) 1992-07-29 1997-06-24 Drymed As Composition comprising fertilized shell eggs
US5286506A (en) 1992-10-29 1994-02-15 Bio-Technical Resources Inhibition of food pathogens by hop acids
US5370863A (en) * 1992-12-16 1994-12-06 Miller Brewing Company Oral care compositions containing hop acids and method
US5296637A (en) 1992-12-31 1994-03-22 Kalamazoo Holdings, Inc. Production of odor-free tetrahydroisohumulates from alpha acids via their tetrahydrohumulates and subsequent isomerization
US5866162A (en) 1993-08-10 1999-02-02 Smithkline Beecham P.L.C. Pharmaceutical composition containing a drug/β-cyclodextrin complex in combination with an acid-base couple
JP2677762B2 (en) * 1994-04-08 1997-11-17 株式会社神戸製鋼所 Oil-cooled compressor
ES2129691T3 (en) 1994-04-12 1999-06-16 Hoechst Marion Roussel Ltd PHARMACEUTICAL COMPOSITION TO TREAT OSTEOPOROSIS.
AT404469B (en) 1994-05-06 1998-11-25 Tulln Zuckerforschung Gmbh METHOD FOR THE PRESERVATION OF SUGAR-BASED PLANT EXTRACTS OR. JUICES
US5523489A (en) * 1995-02-07 1996-06-04 Miller Brewing Company Preparation of tetrahydroisohumulones
IN184685B (en) 1996-02-14 2000-09-23 Nat Inst Immunology
US5827895A (en) 1996-02-27 1998-10-27 Regents Of The University Of Minnesota Hexahydrolupulones useful as anticancer agents
US6020019A (en) 1996-03-26 2000-02-01 Miller Brewing Company Hydrogenation of hop soft resins using CO2
US6589994B1 (en) 1996-08-30 2003-07-08 Nps Pharmaceuticals, Inc. Treating a variety of pathological conditions, including spasticity and convulsions, by effecting a modulation of CNS activity with isovaleramide, isovaleric acid, or a related compound
US5968539A (en) 1997-06-04 1999-10-19 Procter & Gamble Company Mild, rinse-off antimicrobial liquid cleansing compositions which provide residual benefit versus gram negative bacteria
MY120052A (en) 1997-09-17 2005-08-30 Honda Motor Co Ltd Lock-up control device
US6251461B1 (en) 1997-10-10 2001-06-26 S. S. Steiner, Inc. Antimicrobial activity of hops extract against Clostridium botulinum, Clostridium difficile and Helicobacter pylori
WO1999044623A1 (en) 1998-03-04 1999-09-10 Nps Pharmaceuticals, Inc. Compositions comprising valerian extracts, isovaleric acid or derivatives thereof with a nsaid
US6224871B1 (en) 1998-03-11 2001-05-01 Reliv International, Inc. Dietary supplement for nutritionally promoting healthy joint function
US5919813C1 (en) 1998-03-13 2002-01-29 Univ Johns Hopkins Med Use of a protein tyrosine kinase pathway inhibitor in the treatment of diabetic retinopathy
JPH11335231A (en) * 1998-05-20 1999-12-07 Shiseido Co Ltd Skin lotion for slimming body
AU4217499A (en) * 1998-05-29 1999-12-13 Adams Food Ltd. Composition having therapeutic and/or nutritionally active substituent
US7045519B2 (en) 1998-06-19 2006-05-16 Chiron Corporation Inhibitors of glycogen synthase kinase 3
GB2336363B (en) * 1998-08-13 2000-03-15 English Hop Products Limited Hydrogenation of hop acids
DE19841615A1 (en) 1998-09-11 2000-03-16 Fritz Armin Mueller Medicinal wine for alleviating pain and other symptoms of premenstrual syndrome, comprising mixture of extracts of different plants in dry white wine
ES2147538B1 (en) 1999-01-29 2001-04-01 Revlon Consumer Prod Corp A CAPILLARY LOTION WITH IMPROVED PROPERTIES IN ITS HAIR PROTECTIVE AND PREVENTIVE ACTION OF HIS FALL, AND REDUCTION OF THE EXTERNAL EFFECTS OF ANDROGENETIC ALOPECIA AND WITH THAT OF THE HAIR FALL.
ZA200000857B (en) 1999-02-11 2001-05-31 Marx Anna Susan Pharmaceutical composition.
US6801860B1 (en) 1999-02-15 2004-10-05 Genetics Institute, Llc Crystal structure of cPLA2 and methods of identifying agonists and antagonists using same
US6383527B1 (en) 1999-03-04 2002-05-07 Nps Pharmaceuticals, Inc. Compositions comprising valerian extracts, isovaleric acid or derivatives thereof with a NSAID
US6210701B1 (en) 1999-04-30 2001-04-03 Healthcomm International, Inc. Medical food for treating inflammation-related diseases
JP4173606B2 (en) 1999-06-18 2008-10-29 サントリー株式会社 Method for producing low acid beverage
US6129907A (en) * 1999-08-04 2000-10-10 Colgate Palmolive Company Stable hydrogenated lupulone antibacterial oral compositions
WO2001021165A1 (en) 1999-09-21 2001-03-29 Rutgers, The State University Resveratrol analogs for prevention of disease
US6264995B1 (en) 1999-10-19 2001-07-24 Thomas Newmark Herbal composition for reducing inflammation and methods of using same
JP2001161338A (en) 1999-12-09 2001-06-19 Kyodo Shoji:Kk Method for producing sparkling liquor
US6200594B1 (en) 1999-12-29 2001-03-13 Vital Dynamics, Inc. Breast-enhancing, herbal compositions and methods of using same
US6953593B2 (en) 2000-02-01 2005-10-11 Lipoprotein Technologies, Inc. Sustained-release microencapsulated delivery system
US6583322B1 (en) 2000-02-25 2003-06-24 Kalamazoo Holdings, Inc. Dihydro and hexahydro isoalpha acids having a high ratio of trans to cis isomers, production thereof, and products containing the same
US20020086070A1 (en) 2000-03-11 2002-07-04 Kuhrts Eric Hauser Anti-inflammatory and connective tissue repair formulations
CN1427707A (en) 2000-03-31 2003-07-02 日清制油株式会社 External preparation for skin and beautifying agents
US6440465B1 (en) 2000-05-01 2002-08-27 Bioderm, Inc. Topical composition for the treatment of psoriasis and related skin disorders
US6492429B1 (en) 2000-07-10 2002-12-10 N.V. Nutricia Composition for the treatment of osteoarthritis
US20020076452A1 (en) 2000-08-01 2002-06-20 Ashni Naturaceuticals, Inc. Combinations of sesquiterpene lactones and ditepene lactones or triterpenes for synergistic inhibition of cyclooxygenase-2
US6629835B2 (en) * 2000-08-01 2003-10-07 Metaproteomics, Llc Combinations of diterpene triepoxide lactones and ditepene lactones or triterpenes for synergistic inhibition of cyclooxygenase-2
US6908630B2 (en) 2000-08-01 2005-06-21 Metaproteomics, Llc Combinations of sesquiterpene lactones and ditepene triepoxide lactones for synergistic inhibition of cyclooxygenase-2
FR2815227B1 (en) 2000-10-17 2003-04-11 Schwartz Laboratoires Robert ANTI-STRESS COMPOSITION FOR PRIMARY INCORPORATION IN NUTRITIONAL VEHICLES
US6790459B1 (en) 2000-11-03 2004-09-14 Andrx Labs, Llc Methods for treating diabetes via administration of controlled release metformin
JP2002302441A (en) * 2001-02-02 2002-10-18 Nisshin Oil Mills Ltd:The Composition and food and drink for obtunding symptom of premenstrual syndrome
US20030035851A1 (en) 2001-02-08 2003-02-20 Sophie Chen Anti-cancer agents and method of use thereof
US6391346B1 (en) * 2001-04-05 2002-05-21 Thomas Newmark Anti-inflammatory, sleep-promoting herbal composition and method of use
US20030003212A1 (en) 2001-06-13 2003-01-02 Givaudan Sa Taste modifiers
US7901713B2 (en) 2001-06-20 2011-03-08 Metaproteomics, Llc Inhibition of COX-2 and/or 5-LOX activity by fractions isolated or derived from hops
US7901714B2 (en) 2001-06-20 2011-03-08 Metaproteomics, Llp Treatment modalities for autoimmune diseases
US20040219240A1 (en) 2001-06-20 2004-11-04 Babish John G. Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity
US8168234B2 (en) 2001-06-20 2012-05-01 Metaproteomics, Llc Compositions that treat or inhibit pathological conditions associated with inflammatory response
US7718198B2 (en) 2001-06-20 2010-05-18 Metaproteomics, Llc Treatment modalities for autoimmune diseases
US7205151B2 (en) 2001-06-20 2007-04-17 Metaproteomics, Llc Complex mixtures exhibiting selective inhibition of cyclooxygenase-2
US7270835B2 (en) 2001-06-20 2007-09-18 Metaproteomics, Llc Compositions that treat or inhibit pathological conditions associated with inflammatory response
US8142819B2 (en) 2002-10-21 2012-03-27 Metaproteomics, Llc Synergistic compositions that treat or inhibit pathological conditions associated with inflammatory response
US20040115290A1 (en) 2001-06-20 2004-06-17 Tripp Matthew L. Modulation of inflammation by hops fractions and derivatives
US20030096027A1 (en) 2001-10-26 2003-05-22 Babish John G. Curcuminoid compositions exhibiting synergistic inhibition of the expression and/or activity of cyclooxygenase-2
US7279185B2 (en) 2001-10-26 2007-10-09 Metaproteonics, Llc Curcuminoid compositions exhibiting synergistic inhibition of the expression and/or activity of cyclooxygenase-2
KR100993113B1 (en) 2002-02-14 2010-11-08 기린 홀딩스 가부시키가이샤 Compositions and foods for improving lipid metabolism
US7108868B2 (en) 2002-03-22 2006-09-19 Unigen Pharmaceuticals, Inc. Isolation of a dual cox-2 and 5-lipoxygenase inhibitor from acacia
US20030228369A1 (en) 2002-05-06 2003-12-11 Kuhrts Eric Hauser Process for conversion of high viscosity fluids and compositions thereof
NZ539642A (en) 2002-10-21 2007-01-26 Metaproteomics Llc Compositions containing extracts from hops in combination with a second component for treating inflammatory response
US7144590B2 (en) 2003-01-09 2006-12-05 Lipoprotein Technologies, Inc. Bioactive compositions derived from humulus lupulus
EP1543834A1 (en) 2003-12-16 2005-06-22 Biodynamics Production of hop extracts having oestrogenic and antiproliferative bioactivity
US7914831B2 (en) 2004-02-27 2011-03-29 Metaproteomics, Llc Synergistic anti-inflammatory pharmaceutical compositions and related methods using curcuminoids or methylxanthines
US20050192356A1 (en) 2004-02-27 2005-09-01 Babish John G. Synergistic anti-inflammatory pharmaceutical compositions and methods of use
AU2006280074A1 (en) 2005-08-09 2007-02-22 Metaproteomics, Llc Protein kinase modulation by hops and acacia products
US8062898B2 (en) 2006-10-20 2011-11-22 The Board Of Trustees Of The University Of Illinois Selection and rational development of solvent systems in counter-current chromatograph
FR2910325B1 (en) 2006-12-22 2010-03-19 Kronenbourg Brasseries USE OF LUPULONES FOR THE PREVENTION AND THERAPY OF COLORECTAL CANCER.

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110141572A (en) * 2019-06-20 2019-08-20 牡丹江医学院 A kind of drug and preparation method thereof for treating asthma
CN110141572B (en) * 2019-06-20 2020-05-12 牡丹江医学院 Medicine for treating asthma and preparation method thereof

Also Published As

Publication number Publication date
EP1558271A2 (en) 2005-08-03
AU2003286549C1 (en) 2004-05-13
WO2004037180A3 (en) 2004-09-30
AU2003286549A1 (en) 2004-05-13
US8486457B2 (en) 2013-07-16
US20070160692A1 (en) 2007-07-12
AU2003286549B2 (en) 2006-11-30
NZ539642A (en) 2007-01-26
CA2503196A1 (en) 2004-05-06
EP1558271A4 (en) 2006-01-11
WO2004037180A2 (en) 2004-05-06
MXPA05004288A (en) 2005-08-02
JP2006508182A (en) 2006-03-09
KR20050071605A (en) 2005-07-07

Similar Documents

Publication Publication Date Title
CA2503196C (en) Compositions that treat or inhibit pathological conditions associated with inflammatory response
US7431948B2 (en) Compositions that treat or inhibit pathological conditions associated with inflammatory response
US8168234B2 (en) Compositions that treat or inhibit pathological conditions associated with inflammatory response
US7790205B2 (en) Synergistic compositions that treat or inhibit pathological conditions associated with inflammatory response
JP2006508182A5 (en)
US20060127512A1 (en) Modulation of inflammation by hops fractions and derivatives
US20040219240A1 (en) Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity
US7811610B2 (en) Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity
US7807203B2 (en) Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20231020