WO2007149523A2 - Hexahydro-isoalpha acid based protein kinase modulation cancer treatment - Google Patents

Hexahydro-isoalpha acid based protein kinase modulation cancer treatment Download PDF

Info

Publication number
WO2007149523A2
WO2007149523A2 PCT/US2007/014450 US2007014450W WO2007149523A2 WO 2007149523 A2 WO2007149523 A2 WO 2007149523A2 US 2007014450 W US2007014450 W US 2007014450W WO 2007149523 A2 WO2007149523 A2 WO 2007149523A2
Authority
WO
WIPO (PCT)
Prior art keywords
acacia
insulin
cells
adiponectin
hexahydro
Prior art date
Application number
PCT/US2007/014450
Other languages
French (fr)
Other versions
WO2007149523A3 (en
Inventor
Matthew L. Tripp
John G. Babish
Jeffrey Bland
Amy Jennae Hall
Veera Konda
Linda Pacioretty
Anu Desai
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
Application filed by Metaproteomics, Llc filed Critical Metaproteomics, Llc
Priority to JP2009516569A priority Critical patent/JP2009541329A/en
Priority to AU2007261356A priority patent/AU2007261356A1/en
Priority to CA002655059A priority patent/CA2655059A1/en
Priority to EP07796314A priority patent/EP2043621A4/en
Publication of WO2007149523A2 publication Critical patent/WO2007149523A2/en
Publication of WO2007149523A3 publication Critical patent/WO2007149523A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Definitions

  • the present invention relates generally to methods and compositions that can be used to treat or inhibit cancers susceptible to protein kinase modulation. More specifically, the invention relates to methods and compositions which utilize compounds or derivatives commonly isolated either from hops or from members of the plant genus Acacia, or combinations thereof.
  • Signal transduction provides an overarching regulatory mechanism important to maintaining normal homeostasis or, if perturbed, acting as a causative or contributing mechanism associated with numerous disease pathologies and conditions.
  • signal transduction refers to the movement of a signal or signaling moiety from outside of the cell to the cell interior.
  • the signal upon reaching its receptor target, may initiate ligand-receptor interactions requisite to many cellular events, some of which may further act as a subsequent signal.
  • Such interactions serve to not only as a series cascade but moreover an intricate interacting network or web of signal events capable of providing fine-tuned control of homeostatic processes.
  • Signal transducing receptors are generally classified into three classes.
  • the first class of receptors are receptors that penetrate the plasma membrane and have some intrinsic enzymatic activity.
  • Representative receptors that have intrinsic enzymatic activities include those that are tyrosine kinases (e.g. PDGF, insulin, EGF and FGF receptors), tyrosine phosphatases (e.g.
  • CD45 [cluster determinants] protein of T cells and macrophages), guanylate cyclases (e.g. natriuretic peptide receptors) and serine/threonine kinases (e.g. activin and TGF-/3 receptors).
  • Receptors with intrinsic tyrosine kinase activity are capable of autophosphorylation as well as phosphorylation of other substrates.
  • Receptors of the second class are those that are coupled, inside the cell, to
  • GTP-binding and hydrolyzing proteins (termed G-proteins).
  • Receptors of this class which interact with G-proteins have a structure that is characterized by 7 transmembrane spanning domains. These receptors are termed serpentine receptors. Examples of this class are the adrenergic receptors, odorant receptors, and certain hormone receptors (e.g. glucagon, angiotensin, vasopressin and bradykinin).
  • the third class of receptors may be described as receptors that are found intracellularly and, upon ligand binding, migrate to the nucleus where the ligand-receptor complex directly affects gene transcription.
  • the proteins which encode for receptor tyrosine kinases contain four major domains, those being: a) a transmembrane domain, b) an extracellular ligand binding domain, c) an intracellular regulatory domain, and d) an intracellular tyrosine kinase domain.
  • the amino acid sequences of RTKs are highly conserved with those of cAMP-dependent protein kinase (within the ATP and substrate binding regions).
  • RTK proteins are classified into families based upon structural features in their extracellular portions which include the cysteine rich domains, irnmunoglobulin-like domains, cadherin domains, leucine-rich domains, Kringle domains, acidic domains, fibronectin type III repeats, discoidin I-like domains, and EGF-like domains. Based upon the presence of these various extracellular domains the RTKs have been sub-divided into at least 14 different families.
  • SH2 domains proteins that have intrinsic tyrosine kinase activity upon phosphorylation interact with other proteins of the signaling cascade. These other proteins contain a domain of amino acid sequences that are homologous to a domain first identified in the c-Src proto-oncogene. These domains are termed SH2 domains. [009] The interactions of SH2 domain containing proteins with RTKs or receptor associated tyrosine kinases leads to tyrosine phosphorylation of the SH2 containing proteins. The resultant phosphorylation produces an alteration (either positively or negatively) in that activity.
  • SH2 containing proteins that have intrinsic enzymatic activity include phospholipase C- ⁇ (PLC-7), the proto-oncogene c-Ras associated GTPase activating protein (rasGAP), phosphatidylinositol-3 -kinase (PI-3K), protein tyrosine phosphatase- 1C (PTPlC), as well as members of the Src family of protein tyrosine kinases (PTKs).
  • PLC-7 phospholipase C- ⁇
  • rasGAP proto-oncogene c-Ras associated GTPase activating protein
  • PI-3K phosphatidylinositol-3 -kinase
  • PTPlC protein tyrosine phosphatase- 1C
  • PTKs protein tyrosine phosphatase- 1C
  • Non-receptor protein tyrosine kinases by and large couple to cellular receptors that lack enzymatic activity themselves.
  • An example of receptor-signaling through protein interaction involves the insulin receptor (IR).
  • IR insulin receptor
  • This receptor has intrinsic tyrosine kinase activity but does not directly interact, following autophosphorylation, with enzymatically active proteins containing SH2 domains (e.g. PI-3K or PLC- ⁇ ). Instead, the principal IR substrate is a protein termed IRS-I.
  • the receptors for the TGF- ⁇ superfamily represent the prototypical receptor serine/threonine kinase (RSTK).
  • Multifunctional proteins of the TGF- ⁇ superfamily include the activins, inhibins and the bone morphogenetic proteins (BMPs). These proteins can induce and/or inhibit cellular proliferation or differentiation and regulate migration and adhesion of various cell types.
  • BMPs bone morphogenetic proteins
  • TGF-/? is a regulation of progression through the cell cycle.
  • c-Myc one nuclear protein involved in the responses of cells to TGF-/3 is c-Myc, which directly affects the expression of genes harboring Myc-binding elements.
  • PKA, PKC, and MAP kinases represent three major classes of non-receptor serine/threonine kinases.
  • Autoimmune diseases result from a dysfunction of the immune system in which the body produces autoantibodies which attack its own organs, tissues and cells — a process mediated via protein phosphorylation.
  • Over 80 clinically distinct autoimmune diseases have been identified and collectively afflict approximately 24 million people in the US. Autoimmune diseases can affect any tissue or organ of the body. Because of this variability, they can cause a wide range of symptoms and organ injuries, depending upon the site of autoimmune attack. Although treatments exist for many autoimmune diseases, there are no definitive cures for any of them. Treatments to reduce the severity often have adverse side effects.
  • RA Rheumatoid arthritis
  • RA is the most prevalent and best studied of the autoimmune diseases and afflicts about 1% of the population worldwide, and for unknown reasons, like other autoimmune diseases, is increasing.
  • RA is characterized by chronic synovial inflammation resulting in progressive bone and cartilage destruction of the joints.
  • Cytokines, chemokines, and prostaglandins are key mediators of inflammation and can be found in abundance both in the joint and blood of patients with active disease.
  • PGE2 is abundantly present in the synovial fluid of RA patients.
  • Increased PGE2 levels are mediated by the induction of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) at inflamed sites.
  • COX-2 cyclooxygenase-2
  • iNOS inducible nitric oxide synthase
  • the etiology and pathogenesis of RA in humans is still poorly understood, but is viewed to progress in three phases.
  • the initiation phase where dendritic cells present self antigens to autoreactive T cells.
  • the T cells activate autoreactive B cells via cytokines resulting in the production of autoantibodies, which in turn form immune complexes in joints.
  • the immune complexes bind Fcf receptors on macrophages and mast cells, resulting in release of cytokines and chemokines, inflammation and pain.
  • cytokines and chemokines activate and recruit synovial fibroblasts, osteoclasts and polymorphonuclear neutrophils that release proteases, acids, and ROS such as O2-, resulting in irreversible cartilage and bone destruction.
  • Syk is a 72-kDa protein-tyrosine kinase that plays a central role in coupling immune recognition receptors to multiple downstream signaling pathways. This function is a property of both its catalytic activity and its ability to participate in interactions with effector proteins containing SH2 domains. Phosphorylation of Tyr-317, - 342, and -346 create docking sites for multiple SH2 domain containing proteins. [Hutchcroft, J. E., Harrison, M. L. & Geahlen, R. L. (1992). Association of the 72-kDa protein-tyrosine kinase Ptk72 with the B-cell antigen receptor. J. Biol. Chem.
  • Syk has been shown to be required for the activation of PI3K in response to a variety of signals including engagement of the B cell antigen receptor (BCR) and macrophage or neutrophil Fc receptors.
  • BCR B cell antigen receptor
  • macrophage or neutrophil Fc receptors See Crowley, M. T., et al,. J. Exp. Med. 186: 1027- 1039, (1997); Raeder, E. M., et al., J. Immunol. 163, 6785-6793, (1999); and Jiang, K., et al., Blood 101, 236-244, (2003)].
  • the BCR-stimulated activation of PI3K can be accomplished through the phosphorylation of adaptor proteins such as BCAP, CDl 9, or Gabl, which creates binding sites for the p85 regulatory subunit of PI3K.
  • Signals transmitted by many IgG receptors require the activities of both Syk and PI3K and their recruitment to the site of the clustered receptor.
  • PI3K ' with phosphorylated immunoreceptor tyrosine based activation motif sequences on FcgRIIA was proposed as a mechanism for the recruitment of PI3K to the receptor.
  • RIAA Rho isoalpha acid
  • kinases currently being investigated for their association with disease symptomology include Aurora, FGFB, MSK, RSE, and SYK.
  • Aurora - Important regulators of cell division the Aurora family of serine/threonine kinases includes Aurora A, B and C.
  • Aurora A and B kinases have been identified to have direct but distinct roles in mitosis. Over-expression of these three iso forms have been linked to a diverse range of human tumor types, including leukemia, colorectal, breast, prostate, pancreatic, melanoma and cervical cancers.
  • Fibroblast growth factor receptor is a receptor tyrosine kinase.
  • Mutations in this receptor can result in constitutive activation through receptor dimerization, kinase activation, and increased affinity for FGF.
  • FGFR has been implicated in achondroplasia, angiogenesis, and congenital diseases.
  • MSK mitogen- and stress-activated protein kinase 1 and MSK2 are kinases activated downstream of either the ERK (extracellular-signal-regulated kinase) 1/2 or p38 MAPK (mitogen-activated protein kinase) pathways in vivo and are required for the phosphorylation of CREB (cAMP response element-binding protein) and histone H3.
  • Rse is mostly highly expressed in the brain. Rse, also known as Bit, BYK,
  • Dtk, Etk3, Sky, Tif, or sea-related receptor tyrosine kinase is a receptor tyrosine kinase whose primary role is to protect neurons from apoptosis.
  • Rse, AxI, and Mer belong to a newly identified family of cell adhesion molecule-related receptor tyrosine kinases.
  • GAS6 is a Hgand for the tyrosine kinase receptors Rse, AxI, and Mer.
  • GAS6 functions as a physiologic anti-inflammatory agent produced by resting EC and depleted when proinflammatory stimuli turn on the pro-adhesive machinery of EC.
  • Glycogen synthase kinase-3 (GSK-3), present in two isoforms, has been identified as an enzyme involved in the control of glycogen metabolism, and may act as a regulator of cell proliferation and cell death. Unlike many serine-threonine protein kinases, GSK-3 is constitutively active and becomes inhibited in response to insulin or growth factors. Its role in the insulin stimulation of muscle glycogen synthesis makes it an attractive target for therapeutic intervention in diabetes and metabolic syndrome.
  • GSK-3 dysregulation has been shown to be a focal point in the development of insulin resistance. Inhibition of GSK3 improves insulin resistance not only by an increase of glucose disposal rate but also by inhibition of gluconeogenic genes such as phosphoenolpyruvate carboxykinase and glucose-6-phosphatase in hepatocytes. Furthermore, selective GSK3 inhibitors potentiate insulin-dependent activation of glucose transport and utilization in muscle in vitro and in vivo. GSK.3 also directly phosphorylates serine/threonine residues of insulin receptor substrate- 1, which leads to impairment of insulin signaling.
  • GSK3 plays an important role in the insulin signaling pathway and it phosphorylates and inhibits glycogen synthase in the absence of insulin [Parker, P. J., Caudwell, F. B., and Cohen, P. (1983) Eur. J. Biochem. 130:227-234].
  • Increasing evidence supports a negative role of GSK-3 in the regulation of skeletal muscle glucose transport activity.
  • acute treatment of insulin-resistant rodents with selective GSK-3 inhibitors improves whole-body insulin sensitivity and insulin action on muscle glucose transport.
  • Syk is a non-receptor tyrosine kinase related to ZAP-70 involved in signaling from the B-cell receptor and the IgE receptor. Syk binds to ITAM motifs within these receptors, and initiates signaling through the Ras, PI 3-kinase, and PLCg signaling pathways. Syk plays a critical role in intracellular signaling and thus is an important target for inflammatory diseases and respiratory disorders.
  • compositions that act as modulators of kinase can affect a wide variety of disorders in a mammalian body.
  • the instant invention describes compounds and extracts derived from hops or Acacia which may be used to regulate kinase activity, thereby providing a means to treat numerous disease related symptoms with a concomitant increase in the quality of life.
  • the present invention relates generally to methods and compositions that can be used to treat or inhibit cancers susceptible to protein kinase modulation. More specifically, the invention relates to methods and compositions which utilize compounds or derivatives commonly isolated either from hops or from members of the plant genus Acacia, or combinations thereof.
  • a first embodiment of the invention describes methods to treat a cancer responsive to protein kinase modulation in a mammal in need. The method comprises administering to the mammal a therapeutically effective amount of a hexahydro-isoalpha acid.
  • a second embodiment of the invention describes compositions to treat a cancer responsive to protein kinase modulation in a mammal in need where the composition comprises a therapeutically effective amount of a hexahydro-isoalpha acid where the therapeutically effective amount modulates a cancer associated protein kinase.
  • Figure 1 graphically depicts a portion of the kinase network regulating insulin, sensitivity and resistance.
  • Figure 2 graphically depicts the inhibition of five selected kinases by
  • Figure 3 graphically depicts the inhibition of PI3K isoforms by five hops components and a Acacia nilotica extract.
  • Figure 4 depicts RIAA [panel A] and IAA [panel B] dose-related inhibition of
  • Figure 6 provides Western blot detection of COX-2 protein expression.
  • Figure 8 provides a representative schematic of the TransAM NF- ⁇ B kit utilizing a 96-well format.
  • the oligonucleotide bound to the plate contains the consensus binding site for NF- ⁇ B.
  • the primary antibody detected the p50 subunit of NF- ⁇ B.
  • Figure 10 is a schematic of a representative testing procedure for assessing the lipogenic effect of an Acacia sample #4909 extract on developing and mature adipocytes.
  • the 3T3-L1 murine fibroblast model was used to study the potential effects of the test compounds on adipocyte adipogenesis.
  • Figure 11 is a graphic representation depicting the nonpolar lipid content of
  • Figure 12 is a schematic of a representative testing procedure for assessing the effect of a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia sample #4909 on the secretion of adiponectin from insulin-resistant 3T3-L1 adipocytes.
  • Figure 13 is a representative bar graph depicting maximum adiponectin secretion by insulin-resistant 3T3-L1 cells in 24 hr elicited by three doses of troglitazone and four doses of a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia sample #4909. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals.
  • Figure 14 is a schematic of a representative testing protocol for assessing the effect of a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia sample #4909 on the secretion of adiponectin from 3T3-L1 adipocytes treated with test material plus 10, 2 or 0.5 ⁇ g TNF ⁇ /ml.
  • Figure 15 depicts representative bar graphs representing adiponectin secretion by TNF ⁇ treated mature 3T3-L1 cells elicited by indomethacin or an Acacia sample #4909 extract. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals. *Significantly different from TNFo- alone treatment (p ⁇ 0.05).
  • Figure 16 graphically illustrates the relative increase in triglyceride content in insulin resistant 3T3-L1 adipocytes by various compositions of Acacia catechu and A. nilotica from different commercial sources. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals.
  • Figure 17 graphically depicts a representation of the maximum relative adiponectin secretion elicited by various extracts of Acacia catechu. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals.
  • Figure 18 graphically depicts the lipid content (relative to the solvent control) of 3T3-L1 adipocytes treated with hops compounds or the positive controls indomethacin and troglitazone.
  • the 3T3-L1 murine fibroblast model was used to study the potential effects of the test compounds on adipocyte adipogenesis. Results are represented as relative nonpolar lipid content of control cells; error bars represent the 95% confidence interval.
  • Figure 19 is a representative bar graph of maximum adiponectin secretion by insulin-resistant 3T3-L1 cells in 24 hr elicited by the test material over four doses. Values presented are as a percent relative to the solvent control; error bars represent 95% confidence intervals.
  • IAA isoalpha acids
  • RIAA Rho isoalpha acids
  • HHIA hexahydroisoalpha acids
  • THIAA tetrahydroisoalpha acids.
  • Figure 20 depicts the Hofstee plots for Rho isoalpha acids, isoalpha acids, tetrahydroisoalpha acids, hexahydroisoalpha acids, xanthohumols, spent hops, hexahydrocolupulone and the positive control troglitazone.
  • Maximum adiponectin secretion relative to the solvent control was estimated from the y-intercept, while the concentration of test material necessary for half maximal adiponectin secretion was computed from the negative value of the slope.
  • Figure 21 displays two bar graphs representing relative adiponectin secretion by TNF ⁇ -treated, mature 3T3-L1 cells elicited by isoalpha acids and Rho isoalpha acids [panel A], and hexahydro isoalpha acids and tetrahydro isoalpha acids [panel B]. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals. *Significantly different from TNFo; only treatment (p ⁇ 0.05).
  • Figure 22 depicts NF-kB nuclear translocation in insulin-resistant 3T3-L1 adipocytes [panel A] three and [panel B] 24 hr following addition of 10 ng TNFo/ml.
  • Pioglitazone, RIAA and xanthohumols were added at 5.0 (black bars) and 2.5 (stripped bars) ⁇ g/ml.
  • Figure 23 graphically describes the relative triglyceride content of insulin resistant 3T3-L1 cells treated with solvent, metformin, an Acacia sample #5659 aqueous extract or a 1:1 combination of metfo ⁇ min/Acacia catechu extract. Results are represented as a relative triglyceride content of fully differentiated cells in the solvent controls.
  • Figure 24 graphically depicts the effects of 10 ⁇ g/ml of solvent control
  • DMSO dimethyl methoxysulfoxide
  • IAA isoalpha acid
  • THIAA tetrahydroisoalpha acid
  • HHIAA hexahydroisoalpha acid
  • XN xanthohumol
  • LY 249002 LY
  • ETOH ethanol
  • ALPHA alpha acid
  • BETA beta acid
  • Figure 25 graphically depicts the effects of various concentrations of THIAA or reduced isoalpha acids (RIAA) on cell proliferation in the HT-29 cell line.
  • THIAA or reduced isoalpha acids RIAA
  • Figure 26 graphically depicts the effects of various concentrations of THIAA or reduced isoalpha acids (RIAA) on cell proliferation in the SW480 cell line.
  • THIAA or reduced isoalpha acids RIAA
  • Figure 27 graphically depicts the dose responses of various combinations of reduced isoalpha acids (RIAA) and Acacia for reducing serum glucose [panel A] and serum insulin [panel B] in the db/db mouse model.
  • Figure 28 graphically depicts the reduction in serum glucose [panel A] and serum insulin [panel B] in the db/db mouse model produced by a 5:1 combination of RIAA:Acacia as compared to the pharmaceutical anti-diabetic compounds roziglitazone and metformin.
  • RIAA reduced isoalpha acids
  • Figure 29 graphically depicts the effects of reduced isoalpha acids (RIAA) on the arthritic index in a murine model of rheumatoid arthritis.
  • RIAA reduced isoalpha acids
  • FIG. 30 graphically depicts the effects of THIAA on the arthritic index in a murine model of rheumatoid arthritis.
  • Figure 31 graphically summarizes the effects of RIAA and THIAA on collagen induced joint damage.
  • Figure 32 graphically summarizes the effects of RIAA and THIAA on IL-6 levels in a collagen induced arthritis animal model.
  • Figure 33 graphically depicts the effects of RIAA/ 'Acacia (1:5) supplementation (3 tablets per day) on fasting and 2 h post-prandial (pp) insulin levels.
  • pp post-prandial
  • Figure 34 graphically depicts the effects of R ⁇ AA/Acacia (1 :5) supplementation (3 tablets per day) on fasting and 2 h pp glucose levels.
  • 2 h pp glucose level assessment subjects presented after a 10-12 h fast and consumed a solution containing 75 g glucose (Trutol 100, CASCO NERL® Diagnostics); 2 h after the glucose challenge, blood was drawn and assayed for glucose levels (Laboratories Northwest, Tacoma, WA).
  • Figure 35 graphically depicts the effects of BIAA/Acacia (1 :5) supplementation (3 tablets per day) on HOMA scores.
  • HOMA score was calculated from fasting insulin and glucose by published methods [(insulin (mcIU/mL)*glucose (mg/dL))/405].
  • Figure 36 graphically depicts the effects of BJAAJAcacia (1:5) supplementation (3 tablets per day) on serum TG levels.
  • Figure 38 Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
  • Figure 40 Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
  • Figure 44 graphically displays the detection of THIAA in the serum over time following ingestion of 940 mg of THIAA.
  • Figure 45 displays the profile of THIAA detectable in the serum versus contol.
  • Figure 46 depicts the metabolism of THIAA by C YP2C9* 1.
  • the present invention relates generally to methods and compositions that can be used to treat or inhibit cancers susceptible to protein kinase modulation. More specifically, the invention relates to methods and compositions which utilize compounds or derivatives commonly isolated either from hops or from members of the plant genus Acacia, or combinations thereof.
  • the patents, published applications, and scientific literature referred to herein establish the knowledge of those with skill in the art and are hereby incorporated by reference in their entirety to the same extent as if each was specifically and individually indicated to be incorporated by reference. Any conflict between any reference cited herein and the specific teachings of this specification shall be resolved in favor of the latter. Likewise, any conflict between an art-understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this specification shall be resolved in favor of the latter.
  • Standard reference works setting forth the general principles of recombinant DNA technology include Sambrook et ah, Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, New York (1989); Kaufman et al., Eds., Handbook of Molecular and Cellular Methods in Biology in Medicine, CRC Press, Boca Raton (1995); McPherson, Ed., Directed Mutagenesis: A Practical Approach, IRL Press, Oxford (1991). Standard reference works setting forth the general principles of pharmacology include Goodman and Gilman's The Pharmacological Basis of Therapeutics, 1 lth Ed., McGraw Hill Companies Inc., New York (2006).
  • variable can be equal to any integer value of the numerical range, including the end-ooints of the range.
  • the variable can be equal to any real value of the numerical range, including the end-points of the range.
  • a variable which is described as having values between 0 and 2 can be 0, 1 or 2 for variables which are inherently discrete, and can be 0.0, 0.1, 0.01, 0.001, or any other real value for variables which are inherently continuous.
  • a first embodiment of the invention discloses methods to treat a cancer responsive to protein kinase modulation in a mammal in need where the method comprises administering to the mammal a therapeutically effective amount of a hexahydro-isoalpha acid.
  • the hexahydro-isoalpha acid is selected from the group consisting of hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro- adhumulone.
  • the protein kinase modulated is selected from the group consisting of Abl(T315I), Aurora- A, Bmx, CDK9/cyclin Tl, CKI7I, CKl ⁇ 2, CKI73, cSRC, DAPKl, DAPK2, EphBl, ErbB4, Fer, FGFR2, GSK3 ⁇ , GSK3 ⁇ , HBPK3, IGF-IR, MAPKAP-K2, MSK2, PAK3, PAK5, PI3K, Pim-1, PKA(b), PKBB, PKB7, PRAK, Rsk2, Syk, Tie2, TrkA, TrkB, and ZEPK.
  • the cancer responsive to kinase modulation is selected from the group consisting of bladder, breast, cervical, colon, lung, lymphoma, melanoma, prostate, thyroid, and uterine cancer.
  • compositions used in the methods of this embodiment may further comprise one or more members selected from the group consisting of antioxidants, vitamins, minerals, proteins, fats, and carbohydrates, or a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents.
  • a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents.
  • disease associated kinase means those individual protein kinases or groups or families of kinases that are either directly causative of the disease or whose activation is associated with pathways which serve to exacerbate the symptoms of the disease in question.
  • protein kinase modulation is beneficial to the health of the subject refers to those instances wherein the kinase modulation (either up or down regulation) results in reducing, preventing, and/or reversing the symptoms of the disease or augments the activity of a secondary treatment modality.
  • a cancer responsive to protein kinase modulation refers to those instances where administration of the compounds of the invention either a) directly modulates a kinase in the cancer cell where that modulation results in an effect beneficial to the health of the subject (e.g., apoptosis or growth inhibition of the target cancer cell; b) modulates a secondary kinase wherein that modulation cascades or feeds into the modulation of a kinase which produces an effect beneficial to the health of the subject; or c) the target kinases modulated render the cancer cell more susceptible to secondary treatment modalities (e.g., chemotherapy or radiation therapy).
  • secondary treatment modalities e.g., chemotherapy or radiation therapy
  • the terms “comprise(s)” and “comprising” are to be interpreted as having an open- ended meaning. That is, the terms are to be interpreted synonymously with the phrases “having at least” or “including at least”.
  • the term “comprising” means that the process includes at least the recited steps, but may include additional steps.
  • the term “comprising” means that the compound or composition includes at least the recited features or compounds, but may also include additional features or compounds.
  • 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.
  • 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.
  • Spent hops refers to the hops plant products remaining following a hops extraction procedure. See Verzele, M. and De Keukeleire, D., Developments in Food Science 27: Chemistry and Analysis of Hop and Beer Bitter Acids, Elsevier Science Pub. Co., 1991, New York, USA, herein incorporated by reference in its entirety, for a detailed discussion of hops chemistry.
  • Rho refers to those reduced isoalpha acids wherein the reduction is a reduction of the carbonyl group in the 4-methyl-3-pentenoyl side chain.
  • solvent refers to a liquid of aqueous or organic nature possessing the necessary characteristics to extract solid material from the hop plant product.
  • solvents would include, but not limited to, water, steam, superheated water, methanol, ethanol, hexane, chloroform, liquid CO 2 , liquid N 2 or any combinations of such materials.
  • CO 2 extract refers to the solid material resulting from exposing a hops plant product to a liquid or supercritical CO 2 preparation followed by subsequent removal of the CO 2 .
  • compounds may be identified either by their chemical structure, chemical name, or common name. When the chemical structure and chemical or common name conflict, the chemical structure is determinative of the identity of the compound.
  • the compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers.
  • the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated or identified compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
  • Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the skilled artisan.
  • the compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated or identified compounds.
  • the compounds described also encompass isotopically labeled compounds where one or more atoms have an atomic mass different from the atomic mass conventionally found in nature.
  • isotopes that may be incorporated into the compounds of the invention include, but are not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O 5 17 O, etc.
  • Compounds may exist in unsolvated forms as well as solvated forms, including hydrated forms and as N-oxides. In general, compounds may be hydrated, solvated or N-oxides. Certain compounds may exist in multiple crystalline or amorphous forms.
  • congeners, analogs, hydrolysis products, metabolites and precursor or prodrugs of the compound In general, unless otherwise indicated, all physical forms are equivalent for the uses contemplated herein and are intended to be within the scope of the present invention.
  • Compounds according to the invention may be present as salts.
  • pharmaceutically acceptable salts of the compounds are contemplated.
  • a “pharmaceutically acceptable salt” of the invention is a combination of a compound of the invention and either an acid or a base that forms a salt (such as, for example, the magnesium salt, denoted herein as "Mg” or “Mag”) with the compound and is tolerated by a subject under therapeutic conditions.
  • a pharmaceutically acceptable salt of a compound of the invention will have a therapeutic index (the ratio of the lowest toxic dose to the lowest therapeutically effective dose) of 1 or greater. The person skilled in the art will recognize that the lowest therapeutically effective dose will vary from subject to subject and from indication to indication, and will thus adjust accordingly.
  • hop refers to plant cones of the genus Humulus which contain a bitter aromatic oil which is used in the brewing industry to prevent bacterial action and add the characteristic bitter taste to beer. More preferably, the hops used are derived from Humulus lupulus.
  • acacia refers to any member of leguminous trees and shrubs of the genus Acacia.
  • the botanical compound derived from acacia is derived from Acacia catechu or Acacia nilotica.
  • compositions according to the invention are optionally formulated in a pharmaceutically acceptable vehicle with any of the well known pharmaceutically acceptable carriers, including diluents and excipients (see Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, Mack Publishing Co., Easton, PA 1990 and Remington: The Science and Practice of Pharmacy, Lippincott, Williams & Wilkins, 1995). While the type of pharmaceutically acceptable carrier/vehicle employed in generating the compositions of the invention will vary depending upon the mode of administration of the composition to a mammal, generally pharmaceutically acceptable carriers are physiologically inert and nontoxic. Formulations of compositions according to the invention may contain more than one type of compound of the invention), as well any other pharmacologically active ingredient useful for the treatment of the symptom/condition being treated.
  • modulate or “modulation” is used herein to mean the up or down regulation of expression or activity of the enzyme by a compound, ingredient, etc., to which it refers.
  • protein kinase represent transferase class enzymes that are able to transfer a phosphate group from a donor molecule to an amino acid residue of a protein. See Kostich, M., et ah, Human Members of the Eukaryotic Protein Kinase Family, Genome Biology 3(9):research0043.1-0043.12, 2002 herein incorporated by reference in its entirety, for a detailed discussion of protein kinases and family/group nomenclature.
  • kinases include AbI, Abl(T315I),
  • the kinases may be ALK, Aurora- A, AxI, CDK9/cyclin Tl, DAPKl, DAPK2, Fer, FGFR4, GSK3B, GSK3 ⁇ , Hck, JNK2o2, MSK2, p70S6K, PAK3, PI3K delta, PI3K gamma, PKA, PKBB, PKBo, Rse, Rsk2, Syk, TrkA, and TSSKl.
  • the kinase is selected from the group consisting of ABL, AKT, AURORA, CDK, DBF2/20, EGFR, EPH/ELK/ECK, ERK/MAPKFGFR, GSK3, IKKB, BSfSR, JAK DOM 1/2, MARK/PRKAA, MEK/STE7, MEKK/STE11, MLK, mTOR, PAK/STE20, PDGFR, PI3K, PKC, POLO, SRC, TEC/ATK, and ZAP/SYK.
  • compositions of the present invention are intended for use with any mammal that may experience the benefits of the methods of the invention.
  • mammals Foremost among such mammals are humans, although the invention is not intended to be so limited, and is applicable to veterinary uses.
  • "mammals” or “mammal in need” include humans as well as non-human mammals, particularly domesticated animals including, without limitation, cats, dogs, and horses.
  • autoimmune disorder refers to those diseases, illnesses, or conditions engendered when the host's systems are attacked by the host's own immune system.
  • autoimmune diseases include alopecia areata, ankylosing spondylitis, arthritis, antiphospholipid syndrome, autoimmune Addison's disease, autoimmune hemolytic anemia, autoimmune inner ear disease (also known as Meniers disease), autoimmune lymphoproliferative syndrome (ALPS), autoimmune thrombocytopenic purpura, autoimmune hemolytic anemia, autoimmune hepatitis, Bechet's disease, Crohn's disease, diabetes mellitus type 1, glomerulonephritis, Graves' disease, Guillain-Barre syndrome, inflammatory bowel disease, lupus nephritis, multiple sclerosis, myasthenia gravis, pemphigus, pernicious anemia, polyarteritis nodosa, polymyositis,
  • kinases associated with autoimmune disorders include AMPK, BTK, ERK, FGFR 5 FMS, GSK, IGFR, IKK, JAK, PDGFR, PDK, PKC, PLK, ROCK, and VEGFR.
  • Allergic disorders refers to an exaggerated or pathological reaction (as by sneezing, respiratory distress, itching, or skin rashes) to substances, situations, or physical states that are without comparable effect on the average individual.
  • inflammatory disorders means a response (usually local) to cellular injury that is marked by capillary dilatation, leukocytic infiltration, redness, heat, pain, swelling, and often loss of function and that serves as a mechanism initiating the elimination of noxious agents and of damaged tissue.
  • allergic or inflammatory disorders include, without limitation, asthma, rhinitis, ulcerative colitis, Crohn's disease, pancreatitis, gastritis, benign tumors, polyps, hereditary polyposis syndrome, colon cancer, rectal cancer, breast cancer, prostate cancer, stomach cancer, ulcerous disease of the digestive organs, stenocardia, atherosclerosis, myocardial infarction, sequelae of stenocardia or myocardial infarction, senile dementia, and cerebrovascular diseases.
  • kinases associated with allergic disorders include AKT, AMPK, BTK, CHK, EGFR, FYN, IGF-IR, IKKB, ITK, JAK, KIT, LCK, LYN, MAPK, MEK, mTOR, PDGFR, PBK, PKC, PPAR, ROCK, SRC, SYK, and ZAP.
  • metabolic syndrome and "diabetes associated disorders” refers to insulin related disorders, i.e., to those diseases or conditions where the response to insulin is either causative of the disease or has been implicated in the progression or suppression of the disease or condition.
  • insulin related disorders include, without limitation diabetes, diabetic complications, insulin sensitivity, polycystic ovary disease, hyperglycemia, dyslipidemia, insulin resistance, metabolic syndrome, obesity, body weight gain, inflammatory diseases, diseases of the digestive organs, stenocardia, myocardial infarction, sequelae of stenocardia or myocardial infarction, senile dementia, and cerebrovascular dementia.
  • inflammatory conditions include diseases of the digestive organs (such as ulcerative colitis, Crohn's disease, pancreatitis, gastritis, benign tumor of the digestive organs, digestive polyps, hereditary polyposis syndrome, colon cancer, rectal cancer, stomach cancer and ulcerous diseases of the digestive organs), stenocardia, myocardial infarction, sequelae of stenocardia or myocardial infarction, senile dementia, cerebrovascular dementia, immunological diseases and cancer in general.
  • Non-limiting examples of kinases associated with metabolic syndrome can include AKT, AMPK, CDK, CSK, ERK, GSK, IGFR, JNK, MAPK, MEK, PBK, and PKC.
  • Insulin resistance refers to a reduced sensitivity to insulin by the body's insulin-dependent processes resulting in lowered activity of these processes or an increase in insulin production or both. Insulin resistance is typical of type 2 diabetes but may also occur in the absence of diabetes.
  • diabetic complications include, without limitation, retinopathy, muscle infarction, idiopathic skeletal hyperostosis and bone loss, foot ulcers, neuropathy, arteriosclerosis, respiratory autonomic neuropathy and structural derangement of the thorax and lung parenchyma, left ventricular hypertrophy, cardiovascular morbidity, progressive loss of kidney function, and anemia.
  • cancer refers to any of various benign or malignant neoplasms characterized by the proliferation of anaplastic cells that, if malignant, tend to invade surrounding tissue and metastasize to new body sites.
  • Representative, non-limiting examples of cancers considered within the scope of this invention include brain, breast, colon, kidney, leukemia, liver, lung, and prostate cancers.
  • Non-limiting examples of cancer associated protein kinases considered within the scope of this invention include ABL, AKT, AMPK, Aurora, BRK, CDK, CHK, EGFR, ERB, FGFR, IGFR, KIT, MAPK, mTOR, PDGFR, PBK, PKC, and SRC.
  • Ocular disorders refers to those disturbances in the structure or function of the eye resulting from developmental abnormality, disease, injury, age or toxin.
  • Non-limiting examples of ocular disorders considered within the scope of the present invention include retinopathy, macular degeneration or diabetic retinopathy.
  • Ocular disorder associated kinases include, without limitation, AMPK, Aurora, EPH, ERB, ERK, FMS, IGFR, MEK, PDGFR, PI3K, PKC, SRC, and VEGFR.
  • a "neurological disorder”, as used herein, refers to any disturbance in the structure or function of the central nervous system resulting from developmental abnormality, disease, injury or toxin.
  • Representative, non-limiting examples of neurological disorders include Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS or Lou Gehrig's Disease), Huntington's disease, neurocognitive dysfunction, senile dementia, and mood disorder diseases.
  • Protein kinases associated with neurological disorders may include, without limitation, AMPK, CDK, FYN, JNK, MAPK, PKC, ROCK, RTK, SRC, and VEGFR.
  • Cardiovascular disease refers to those pathologies or conditions which impair the function of, or destroy cardiac tissue or blood vessels.
  • Cardiovascular disease associated kinases include, without limitation, AKT, AMPK, GRK 5 GSK, IGF-IR, DCKB, JAK, JUN, MAPK, PKC, RHO, ROCK, and TOR.
  • Osteoporosis refers to a disease in which the bones have become extremely porous, thereby making the bone more susceptible to fracture and slower healing.
  • Protein kinases associated with osteoporosis include, without limitation, AKT, AMPK, CAMK, IRAK-M, MAPK, mTOR, PPAR, RHO, ROS, SRC, SYR, and VEGFR.
  • compositions to treat a cancer responsive to protein kinase modulation in a mammal in need comprise a therapeutically effective amount of a hexahydro-isoalpha acid; wherein the therapeutically effective amount modulates a cancer associated protein kinase.
  • the hexahydro-isoalpha acid is selected from the group consisting of hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.
  • compositions further comprise a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents.
  • a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents.
  • the compositions further comprise one or more members selected from the group consisting of antioxidants, vitamins, minerals, proteins, fats, and carbohydrates.
  • treating is meant reducing, preventing, and/or reversing the symptoms in the individual to which a compound of the invention has been administered, as compared to the symptoms of an individual not being treated according to the invention.
  • a practitioner will appreciate that the compounds, compositions, and methods described herein are to be used in concomitance with continuous clinical evaluations by a skilled practitioner (physician or veterinarian) to determine subsequent therapy. Hence, following treatment the practitioners will evaluate any improvement in the treatment of the pulmonary inflammation according to standard methodologies. Such evaluation will aid and inform in evaluating whether to increase, reduce or continue a particular treatment dose, mode of administration, etc.
  • a compound of the invention may be administered prophylactically, prior to any development of symptoms.
  • the term "therapeutic,” “therapeutically,” and permutations of these terms are used to encompass therapeutic, palliative as well as prophylactic uses.
  • by “treating or alleviating the symptoms” is meant reducing, preventing, and/or reversing the symptoms of the individual to which a compound of the invention has been administered, as compared to the symptoms of an individual receiving no such administration.
  • therapeutically effective amount is used to denote treatments at dosages effective to achieve the therapeutic result sought.
  • therapeutically effective amount of the compound of the invention may be lowered or increased by fine tuning and/or by administering more than one compound of the invention, or by administering a compound of the invention with another compound. See, for example, Meiner, C.L., “Clinical Trials: Design, Conduct, and Analysis,” Monographs in Epidemiology and Biostatistics, Vol. 8 Oxford University Press, USA (1986).
  • the invention therefore provides a method to tailor the administration/treatment to the particular exigencies specific to a given mammal.
  • therapeutically effective amounts may be easily determined for example empirically by starting at relatively low amounts and by step-wise increments with concurrent evaluation of beneficial effect.
  • number of administrations of the compounds according to the invention will vary from patient to patient based on the particular medical status of that patient at any given time including other clinical factors such as age, weight and condition of the mammal and the route of administration chosen.
  • symptom denotes any sensation or change in bodily function that is experienced by a patient and is associated with a particular disease, i.e., anything that accompanies “X” and is regarded as an indication of "X'”s existence. It is recognized and understood that symptoms will vary from disease to disease or condition to condition.
  • symptoms associated with autoimmune disorders include fatigue, dizziness, malaise, increase in size of an organ or tissue (for example, thyroid enlargement in Grave's Disease), or destruction of an organ or tissue resulting in decreased functioning of an organ or tissue (for example, the islet cells of the pancreas are destroyed in diabetes).
  • Representative symptomology for allergy associated diseases or conditions include absentmindedness, anaphylaxis, asthma, burning eyes, constipation, coughing, dark circles under or around the eyes, dermatitis, depression, diarrhea, difficulty swallowing, distraction or difficulty with concentration, dizziness, eczema, embarrassment, fatigue, flushing, headaches, heart palpitations, hives, impaired sense of smell, irritability/behavioral problems, itchy nose or skin or throat, joint aches muscle pains, nasal congestion, nasal polyps, nausea, postnasal drainage (postnasal drip), rapid pulse, rhinorrhea (runny nose), ringing - popping or fullness in the ears, shortness of breath, skin rashes, sleep difficulties, sneezing, swelling (angioedema), throat hoarseness, tingling nose, tiredness, vertigo, vomiting, watery or itchy or crusty or red eyes, and wheezing.
  • inflammation refers to a local response to cellular injury that is marked by capillary dilatation, leukocytic infiltration, redness, heat, pain, swelling, and often loss of function and that serves as a mechanism initiating the elimination of noxious agents and of damaged tissue.
  • Representative symptoms of inflammation or an inflammatory condition include, if confined to a joint, redness, swollen joint that's warm to touch, joint pain and stiffness, and loss of joint function.
  • Systemic inflammatory responses can produce "flu-like" symptoms, such as, for instance, fever, chills, fatigue/loss of energy, headaches, loss of appetite, and muscle stiffness.
  • Diabetes and metabolic syndrome often go undiagnosed because many of their symptoms seem so harmless. For example, some diabetes symptoms include, without limitation: frequent urination, excessive thirst, extreme hunger, unusual weight loss, increased fatigue, irritability, and blurry vision.
  • Symptomology of neurological disorders may be variable and can include, without limitation, numbness, tingling, hyperesthesia (increased sensitivity), paralysis, localized weakness, dysarthria (difficult speech), aphasia (inability to speak), dysphagia (difficulty swallowing), diplopia (double vision), cognition issues (inability to concentrate, for example), memory loss, amaurosis fugax (temporary loss of vision in one eye) difficulty walking, incoordination, tremor, seizures, confusion, lethargy, dementia, delirium and coma.
  • kinases represent transferase class enzymes that are able to transfer a phosphate group from a donor molecule (usually ATP) to an amino acid residue of a protein (usually threonine, serine or tyrosine).
  • a donor molecule usually ATP
  • an amino acid residue of a protein usually threonine, serine or tyrosine.
  • Kinases are used in signal transduction for the regulation of enzymes, i.e., they can inhibit or activate the activity of an enzyme, such as in cholesterol biosynthesis, amino acid transformations, or glycogen turnover. While most kinases are specialized to a single kind of amino acid residue, some kinases exhibit dual activity in that they can phosphorylate two different kinds of amino acids. As shown in Figure 1, kinases function in signal transduction and translation.
  • hops compounds tested inhibited 25 of the 205 kinases by 10% or greater. Eight (8) of the 205 were inhibited by >20%; 5 of 205 were inhibited by >30; and 2 were inhibited by about 50%.
  • hops inhibits PI3K ⁇ , PI3K ⁇ , PI3K ⁇ ,
  • Aktl Akt2, GSK3 ⁇ , GSK3 ⁇ , P70S6K. It should be noted that mTOR was not available for testing.
  • Aktl null mice are viable, but retarded in growth [Cho et al., Science 292:1728-1731 (2001)]. Drosophila eye cells deficient in Aktl are reduced in size [Verdu et al., Nat cell Biol 1 :500-505 (1999)]; overexpression leads to increased size from normal. Akt2 null mice are viable but have impaired glucose control [Cho et al., J Biol Chem 276:38345-38352 (2001)].
  • Aktl plays a role in size determination and Akt2 is involved in insulin signaling.
  • the PDK pathway is known to play a key role in mRNA stability and mRNA translation selection resulting in differential protein expression of various oncogene proteins and inflammatory pathway proteins.
  • a particular 5' mRNA structure denoted 5'-TOP has been shown to be a key structure in the regulation of mRNA translation selection.
  • hops compounds inhibit cPLA2 protein expression (Western blots, data not shown) but not mRNA, suggests that the anti-inflammatory mode of action of hops compounds may be via reducing substrate availability to COX2 by reducing cPLA2 protein levels, and perhaps more specifically, by inhibiting the PI3K pathway resulting in the inhibition of activation of TOP mRNA translation.
  • IAA isoalpha acids
  • HHIAA heaxahydroisoalpha acids
  • beta acids beta acids
  • xanthohumol xanthohumol
  • Dose response effect (as % of ControD of xanthohumol on selected protein kinases
  • PI3K ⁇ a kinase strongly implicated in autoimmune diseases such as, for example, rheumatoid arthritis and lupus erythematosus, exhibited a response inhibiting 36%, 78% and 87% of kinase activity at 10, 50, and 100 ug/ml respectively for MgRho.
  • MgRho inhibited Syk in a dose dependent manner with 21%, 54% and 72% inhibition at 10, 50, and 100 ⁇ g/ml respectively.
  • GSK or glycogen synthase kinase displayed inhibition following mgRho exposure (alpha, 35, 36, 87% inhibition; beta, 35, 83, 74 % inhibition respectively at 10, 50, 100 ⁇ g/mi). See Table 2.
  • THIAA displayed a dose dependent inhibition of kinase activity for many of the kinases examined with inhibition of FGFR2 of 7%, 16%, 77%, and 91% at 1, 5, 25, and 50 ⁇ g/ml respectively. Similar results were observed for FGFR3 (0%, 6%, 61%, and 84%) and TrkA (24%, 45%, 93%, and 94%) at 1, 5, 25, and 50 ⁇ g/ml respectively. See Table 3.
  • the acacia extract tested appeared to be the most potent inhibitor of kinase activity examined (Table 4), demonstrating 80% or greater inhibition of activity for such kinases as Syk (98%), Lyn (96%), GSK3 ⁇ (95%), Aurora-A (92%), Flt4 (88%), MSSKl (88%), GSK3B (87%), BTK (85%), PRAK (82%), and TrkA (80%), all at a 1 ⁇ g/ml exposure.
  • hops compounds tested showed >50% inhibition of PI3K activity with Mg-THIAA producing the greatest overall inhibition (>80% inhibition for all PI3K isoforms tested). Further note that both xanthohumol and Mg-beta acids were more inhibitory to PI3K- ⁇ than to PI3K-0 or PI3K-6. Mg-IAA was approximately 3-fold more inhibitory to PI3K-/? than to PI3K- ⁇ or PDK- ⁇ . The Acacia nilotica heartwood extract appeared to stimulate PI3K-/3 or PI3K- ⁇ activity. Comparable results were obtained for Syk and GSK kinases (data not shown).
  • the objective of this example was to assess the extent to which hops derivatives inhibited COX-2 synthesis of PGE 2 preferentially over COX-I synthesis of PGE 2 in the murine RAW 264.7 macrophage model.
  • the RAW 264.7 cell line is a well-established model for assessing anti-inflammatory activity of test agents. Stimulation of RAW 264.7 cells with bacterial lipopolysaccharide induces the expression of COX-2 and production of PGE 2 . Inhibition of PGE2 synthesis is used as a metric for anti-inflammatory activity of the test agent.. Equipment, Chemicals and Reagents, PGE 2 assay, and calculations are described below.
  • Equipment - Equipment used in this example included an OHAS Model
  • 055 :B5 was from Sigma (St. Louis, MO). Heat inactivated Fetal Bovine Serum (FBS-HI Cat. #35-011CV), and Dulbecco's Modification of Eagle's Medium (DMEM Cat #10- 013CV) was purchased from Mediatech (Herndon, VA).
  • FBS-HI Cat. #35-011CV Heat inactivated Fetal Bovine Serum
  • DMEM Cat #10- 013CV Dulbecco's Modification of Eagle's Medium
  • 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), (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 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.
  • Test materials Hops derivatives as described in Table 12 were used. The
  • COX-I selective inhibitor aspirin and COX-2 selective inhibitor celecoxib were used as positive controls.
  • Aspirin was obtained from Sigma (St. Louis, MO) and the commercial formulation of celecoxib was used (CelebrexTM, Searle & Co., Chicago, IL).
  • Cell culture and treatment with test material 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.
  • DMEM Dulbecco's Modification of Eagle's Medium
  • 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.
  • PGE 2 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 PGE 2 standard samples, were mixed with appropriate amounts of acetylcholinesterase-labeled tracer and PGE 2 antiserum, and incubated at room temperature for 18 h. After the wells were emptied and rinsed with wash buffer, 200 ⁇ l of Ellman's reagent containing substrate for acetylcholinesterase were added.
  • the reaction was maintained on a slow shaker at room temperature for 1 h and the absorbance at 415 run was determined in a Bio-Tek Instruments (Model #Elx800, Winooski, VT) ELISA plate reader.
  • the PGE 2 concentration was represented as picograms per ml.
  • the manufacturer's specifications for this assay include an intra-assay coefficient of variation of ⁇ 10%, cross reactivity with PGD 2 and PGF 2 of less than 1% and linearity over the range of 10 - 1000 pg ml ' l.
  • the median inhibitory concentrations (IC 50 ) for PGE 2 synthesis from both COX-2 and COX-I were calculated as described below.
  • COX-2 selectivity For extrapolating in vitro data to clinical efficacy, it is generally assumed that a COX-2 selectivity of 5-fold or greater indicates the potential for clinically significant protection of gastric mucosa. Under this criterion, beta acids, CO 2 hop extract, spent hops CCVethanol, tetrahydro isoalpha acids and hexahydro isoalpha acids displayed potentially clinically relevant COX-2 selectivity.
  • test materials were added followed 60 minutes later by the addition of the calcium ionophore A23187.
  • Test materials were dissolved in 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 ⁇ l of this solution was subsequently added to eight wells for each dose of test material.
  • Supernatant media was sampled for PGE 2 determination after 30 minutes. Median inhibitory concentrations were computed from a minimum of four concentrations over two independent experiments as described in Example 4.
  • Cell viability was assessed by microscopic inspection of cells prior to or immediately following sampling of the medium for PGE 2 assay. No apparent cell mortality was noted at any of the concentrations tested.
  • Results - LPS-stimulation of PGE 2 production in RAW 264.7 cells ranged from 1.4-fold to 2.1 -fold relative to non-stimulated cells.
  • RIAA and IAA produced only modest, dose-related inhibition Of PGE 2 .
  • 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 IC 50 values (Table 10) in the mg/ml range for RIAA (36 mg/ml) and IAA (>1000 mg/ml).
  • the minimal changes observed in response over three-log units of doses suggests that the observed PGE 2 inhibitory effect of the hops derivatives in this cell-based assay may be a secondary effect on the cells and not a direct inhibition of COX-2 enzyme activity.
  • Figure 4A and 4B depict the dose-response data respectively, for RIAA and
  • hop materials were among the most active, antiinflammatory natural products tested as assessed by their ability to inhibit PGE 2 biosynthesis in vitro; (2) RIAA and IAA do not appear to be direct COX-2 enzyme inhibitors based on their pattern of inhibition with respect to COX-2 induction; and (3) RIAA and IAA have a COX-2 selectively that appears to be based on inhibition of COX-2 expression, not COX-2 enzyme inhibition. This selectivity differs from celecoxib, whose selectivity is based on differential enzyme inhibition.
  • 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 PGE 2 determination after 30 minutes. Median inhibitory concentrations were computed from a minimum of eight replicates at four concentrations over two independent experiments.
  • Hops compounds and derivatives are not direct cvclooxygenase enzyme inhibitors in A549 pulmonary epithelial cells
  • the cells were routinely cultured at 37°C with 5% CO 2 in RPMI 1640 containing 10% FBS, with 50 units penicillin/ml, 50 ⁇ g streptomycin/ml, 5 mM sodium pyruvate, and 5 mM L-glutamine. On the day of the experiments, exponentially growing cells were harvested and washed with serum-free RPMI 1640.
  • interleukin-l ⁇ (10 ng/ml) was added to induce the expression of COX-2.
  • the cells were washed with serum-free RPMI 1640.
  • 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 arachadonic acid.
  • Results At the doses tested, the experimental protocol failed to capture a median effective concentration for any of the hops extracts or derivatives. Since the protocol requires the stimulation of COX-2 expression prior to the addition of the test compounds, it is believed that the failure of the test materials to inhibit PGE 2 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 was observed using the WHMA-COX-2 protocol, this procedure appears inappropriate in evaluating the anti-inflammatory properties of hops compounds or derivatives of hops compounds.
  • 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), used in this example were previously described in Example 1. All other chemicals were obtained from suppliers as described in Example 4. Test materials at a final concentration of 10 ⁇ g/ml were added 60 minutes prior to the addition of the mite dust allergen.
  • Mite dust allergen isolation - Dermatophagoides farinae is the American house dust mite. D. farinae were raised 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.
  • mite powder 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.
  • Example 6 (American Type Culture Collection, Bethesda, MD) was cultured and treated as previously described in Example 6. Mite allergen was added to the culture medium to achieve a final concentration of 1000 ng/ml. Eighteen hours later, the media were sampled for PGE 2 determination.
  • Results - Table 11 depicts the extent of inhibition by hops derivatives OfPGE 2 biosynthesis in A549 pulmonary cells stimulated by mite dust allergen. All hops derivatives tested were capable of significantly inhibiting the stimulatory effects of mite dust allergens.
  • PGE 2 stimulatory effects of mite dust allergens in A549 pulmonary cells.
  • Cell Culture The murine macrophage RAW 264.7 cell line was purchased from ATCC (Manassas, VA) and maintained according to their instructions. Cells were subcultured in 96-well plates at a density of 8x 10 4 cells per well and allowed to reach 90% confluence, approximately 2 days. LPS (1 ⁇ g /ml) or PBS alone was added to the cell media and incubated for 12 hrs.
  • the media was removed from the wells and LPS (1 ⁇ g/ml) with the test compounds dissolved in DMSO and serum-free RPMI, were added to the wells to achieve final concentrations of MgRIAA at 20, 5.0, 1.0 and 0.1 ⁇ g/ml and celecoxib at 100, 10, 1 and 0.1 ng/ml. Each concentration was run in 8 duplicates. Following 1 hr of incubation with the test compounds, the cell media were removed and replaced with fresh media with test compounds with LPS (1 ⁇ g/ml) and incubated for 1 hr. The media were removed from the wells and analyzed for the PGE 2 synthesis.
  • PGE 2 was employed (Cayman Chemical, Ann Arbor, MI). Samples were diluted 10 times in ElA buffer and the recommended procedure of the manufacturer was used without modification. The PGE 2 concentration was represented as picograms per ml. The manufacturer's specifications for this assay include an intra-assay coefficient of variation of ⁇ 10%, cross reactivity with PGD2 and PGF 2 of less than 1% and linearity over the range of 10 - lOOO pg ml "1 .
  • Cell Culture The murine macrophage RAW 264.7 cell line was purchased from ATCC (Manassas, VA) and maintained according to their instructions. Cells were grown and subcultured in 24- well plates at a density of 3 x 10 5 cells per well and allowed to reach 90% confluence, approximately 2 days. Test compounds were added to the cells in serum free medium at a final concentration of 0.4% DMSO. Following 1 hr of incubation with the test compounds, LPS (1 ⁇ g/ml) or phosphate buffered saline alone was added to the cell wells and incubation continued for the indicated times.
  • ATCC Manassas, VA
  • Cell extracts 50 ⁇ g were electrophoresed through a pre-cast 4%-20% Tris-HCl Criterion gel (Bio-Rad, Hercules, CA) until the front migration dye reached 5 mm from the bottom of the gel.
  • the proteins were transferred to nitrocellulose membrane using a semi-dry system from Bio-Rad (Hercules, CA). The membrane was washed and blocked with 5% dried milk powder for 1 hour at room temperature. Incubation with the primary antibody followed by the secondary antibody was each for one hour at room temperature.
  • Chemiluminescence was performed using the SuperSignal West Femto Maximum Sensitivity Substrate from Pierce Biotechnology (Rockford, IL) by incubation of equal volume of luminol/enhancer solution and stable peroxide solution for 5 minutes at room temperature.
  • the Western blot image was captured using a cooled CCD Kodak ® (Rochester, NY) ISlOOO imaging system. Densitometry was performed using Kodak ® software. [00193] The percent of COX-2 and iNOS protein expression was assessed using
  • Test compounds were prepared in dimethyl sulfoxide (DMSO) and stored at -20 0 C.
  • MgRIAA was supplied by Metagenics (San Clemente, CA).
  • Parthenolide, a specific inhibitor for NF-kB activation was purchased from Sigma-Aldrich (St. Louis, MO).
  • the PI3K inhibitor LY294002 was purchased from EMD Biosciences (San Diego, CA).
  • Cell Culture The murine macrophage RAW 264.7 cell line was purchased from ATCC (Manassas, VA) and maintained according to their instructions. Cells were subcultured in 6-well plates at a density of 1.5 x 10 6 cells per well and allowed to reach 90% confluence, approximately 2 days. Test compounds MgRIAA (55 and 14 ⁇ g/ml), parthenolide (80 ⁇ M) and LY294002 (25 ⁇ M) were added to the cells in serum free media at a final concentration of 0.4% DMSO. Following 1 hr of incubation with the test compounds, LPS (1 ⁇ g/ml) or PBS alone was added to the cell media and incubation continued for an additional four hours.
  • MgRIAA 55 and 14 ⁇ g/ml
  • parthenolide 80 ⁇ M
  • LY294002 25 ⁇ M
  • NF-KB-DNA binding - Nuclear extracts were prepared essentially as described by Dignam, et al [Nucl Acids Res 11:1475-1489, (1983)]. Briefly, cells were washed twice with cold PBS, then Buffer A (10 mM HEPES, pH 7.0; 1.5 mM MgCl 2 ; 10 mM KCl; 0.1% NP-40; aprotinin 5 ⁇ g/ml; pepstatin A 1 ⁇ g/ml; leupeptin 5 ⁇ g/ml; phenylmethanesulfonyl fluoride 1 mM) was added and allowed to sit on ice for 15 minutes.
  • Buffer A (10 mM HEPES, pH 7.0; 1.5 mM MgCl 2 ; 10 mM KCl; 0.1% NP-40; aprotinin 5 ⁇ g/ml; pepstatin A 1 ⁇ g/ml; leupeptin 5 ⁇ g/ml;
  • the nuclear extract fraction was collected as the supernatant layer following centrifugation at 10,000 x g for 5 min at 4°C.
  • NF-kB DNA binding of the nuclear extracts was assessed using the TransAM NF- ⁇ B kit from Active Motif (Carlsbad, CA) as per manufacturer's instructions.
  • the TransAM kit detected the p50 subunit of NF- ⁇ B binding to the consensus sequence in a 96-well format. Protein concentration was measured (Bio-Rad assay) and 10 ⁇ g of nuclear protein extracts were assayed in duplicate.
  • MgDHIAA may result in decreased COX-2 protein expression, ultimately leading to a decrease in PGE 2 production.
  • the Model - The 3T3-L1 murine fibroblast model is used to study the potential effects of compounds on adipocyte differentiation and adipogenesis.
  • This cell line allows investigation of stimuli and mechanisms that regulate preadipocytes replication separately from those that regulate differentiation to adipocytes [Fasshauer, M., Klein, J., Neumann, S., Eszlinger, M., and Paschke, R. Hormonal regulation of adiponectin gene expression in 3T3- Ll adipocytes. Biochem Biophys Res Commun, 290: 1084-1089, (2002); Li, Y. and Lazar, M. A.
  • 3T3-L1 cells As preadipocytes, 3T3-L1 cells have a fibroblastic appearance. They replicate in culture until they form a confluent monolayer, after which cell-cell contact triggers Go/G] growth arrest. Terminal differentiation of 3T3-L1 cells to adipocytes depends on proliferation of both pre- and post-confluent preadipocytes. Subsequent stimulation with 3- isobutyl-1-methylxanthane, dexamethasone, and high does of insulin (MDI) for two days prompts these cells to undergo post-confluent mitotic clonal expansion, exit the cell cycle, and begin to express adipocyte-specific genes.
  • MDI isobutyl-1-methylxanthane
  • MDI high does of insulin
  • Thiazolidinediones such as troglitazone and pioglitazone have been shown to selectively stimulate lipogenic activities in fat cells resulting in greater insulin suppression of lipolysis or release of fatty acids into the plasma [Yamauchi, T., J. Kamon, et al.
  • PPARgamma peroxisome proliferator-activated receptor gamma
  • Thiazolidinediones increase plasma-adipose tissue FFA exchange capacity and enhance insulin-mediated control of systemic FFA availability. Diabetes 50(5): 1158-65, (2001)]. This action would leave less free fatty acids available for other tissues [Yang, W. S., W. J. Lee, et al. Weight reduction increases plasma levels of an adipose-derived antiinflammatory protein, adiponectin. J Clin Endocrinol Metab 86(8): 3815-9, (2001)]. Thus, insulin desensitizing effects of free fatty acids in muscle and liver would be reduced as a consequence of thiazolidinedione treatment. These in vitro results have been confirmed clinically [Boden, G.
  • Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS-HI (fetal bovine serum-heat inactivated) from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
  • the murine fibroblast cell line 3T3-L1 was purchased from the American Type Culture Collection (Manassas, VA) and sub-cultured according to instructions from the supplier. Prior to experiments, cells were cultured in DMEM containing 10% FBS-HI added 50 units penicillin/ml and 50 ⁇ g streptomycin/ml, and maintained in log phase prior to experimental setup. Cells were grown in a 5% CO 2 humidified incubator at 37°C. Components of the pre-confluent medium included (1) 10% FBS/DMEM containing 4.5 g glucose/L; (2) 50 U/ml penicillin; and (3) 50 ⁇ g/ml streptomycin.
  • Growth medium was made by adding 50 ml of heat inactivated FBS and 5 ml of penicillin/streptomycin to 500 ml DMEM. This medium was stored at 4 ° C. Before use, the medium was warmed to 37 ° C in a water bath.
  • 3T3-TI cells were seeded at an initial density of 6x10 4 cells/cm 2 in 24-well plates. For two days, the cells were allowed grow to reach confluence. Following confluence, the cells were forced to differentiate into adipocytes by the addition of differentiation medium; this medium consisted of (1) 10% FBS/DMEM (high glucose); (2) 0.5 mM methylisobutylxanthine; (3) 0.5 ⁇ M dexamethasone and (4) 10 ⁇ g/ml insulin (MDI medium). After three days, the medium was changed to post-differentiation medium consisting of 10 ⁇ g/ml insulin in 10% FBS/DMEM.
  • FBS/DMEM high glucose
  • MDI medium 10 ⁇ g/ml insulin
  • Oil Red O Staining - Triglyceride content of D6/D7-differentiated 3T3-L1 cells was estimated with Oil Red O according to the method of Kasturi and Joshi [Kasturi, R. and Joshi, V. C. Hormonal regulation of stearoyl coenzyme A desaturase activity and lipogenesis during adipose conversion of 3T3-L1 cells. J Biol Chem, 257: 12224-12230, 1982]. Monolayer cells were washed with PBS (phosphate buffered saline, Mediatech) and fixed with 10% formaldehyde for ten minutes.
  • PBS phosphate buffered saline, Mediatech
  • BODIPY Staining - 4,4-Difluoro-l ,3,5,7,8-penta-methyl-4-bora-3a,4a-diaza-s- indacene (BODIPY 493/503; Molecular Probes, Eugene, OR) was used for quantification of cellular neutral and nonpolar lipids. Briefly, media were removed and cells were washed once with non-sterile PBS. A stock IOOOX BODIPY/DMSO solution was made by dissolving 1 mg BODIPY in 1 ml DMSO (1,000 ⁇ g BODIPY/ml).
  • a working BODEPY solution was then made by adding 10 ⁇ l of the stock solution to 990 ⁇ l PBS for a final BODIP Y concentration in the working solution of 0.01 ⁇ g/ ⁇ l.
  • One-hundred ⁇ l of this working solution (1 ⁇ g BODIPY) was added to each well of a 96-well microtiter plate. After 15 min on an orbital shaker (DS-500, VWR Scientific Products, South Plainfield, NJ) at ambient temperature, the cells were washed with 100 ⁇ l PBS followed by the addition of 100 ⁇ l PBS for reading for spectrofluorometric determination of BODIPY incorporation into the cells.
  • a Packard Fluorocount spectrofluorometer (Model#BF 10000, Meridan, CT) set at 485 nm excitation and 530 nm emission was used for quantification of BODIPY fluorescence. Results for test materials, indomethacin, and troglitazone were reported relative to the fluorescence of the solvent controls.
  • Results The positive controls indomethacin and troglitazone induced lipogenesis to a similar extent in 3T3-L1 cells ( Figure 11). Unexpectedly, the AcE produced an adipogenic response greater than either of the positive controls indomethacin and troglitazone.
  • Example 12 Increased adiponectin secretion from insulin-resistant 3T3-L1 adipocytes elicited by a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia.
  • Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS-HI (fetal bovine serum-heat inactivated from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
  • the Acacia extract was tested at 50, 25, 12.5 and 6.25 ⁇ g/ml. Twenty-four hours later, the supernatant medium was sampled for adiponectin determination. The complete procedure for differentiation and treatment of cells with test materials is outlined schematically in Figure 12.
  • Adiponectin Assay The adiponectin secreted into the medium was quantified using the Mouse Adiponectin Quantiki ⁇ e® Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN)- Information supplied by the manufacturer indicated that recovery of adiponectin spiked in mouse cell culture media averaged 103% and the minimum detectable adiponectin concentration ranged from 0.001 to 0.007 ng/ml.
  • 3T3-L1 cells, Acacia, and/or apecatechin may be expected to have a positive effect on clinical pathologies in which plasma adiponectin concentrations are depressed.
  • Test Materials Indomethacin, methylisobutylxanthine, dexamethasone, and insulin were obtained from Sigma (St. Louis, MO).
  • the test material was a dark brown powder produced from a 50:50 (v/v) water/alcohol extract of the gum resin of Acacia sample #4909 and was obtained from Bayir Chemicals (No. 68, South Cross Road, Basavanagudi, India). The extract was standardized to contain not less than 20% apecatechin.
  • Batch No. A Cat/2304 used in this example contained 20.8% apecatechin as determined by UV analysis.
  • Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS (fetal bovine serum) characterized from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
  • the medium was changed to post-differentiation medium consisting of 10% FBS in DMEM.
  • the medium was changed to test medium containing 10, 2 or 0.5 ng TNFo/ml in 10% FBS/DMEM with or without indomethacin or Acacia extract.
  • Indomethacin was dissolved in dimethyl sulfoxide and added to achieve concentrations of 5, 2.5, 1.25 and 0.625 ⁇ g/ml.
  • the Acacia extract was tested at 50, 25, 12.5 and 6.25 ⁇ g/ml.
  • the supernatant medium was sampled for adiponectin determination.
  • the complete procedure for differentiation and treatment of cells with test materials is outlined schematically in Figure 14.
  • Adiponectin Assay The adiponectin secreted into the medium was quantified using the Mouse Adiponectin Quantikine® Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN). Information supplied by the manufacturer indicated that recovery of adiponectin spiked in mouse cell culture media averaged 103% and the minimum detectable adiponectin concentration ranged from 0.001 to 0.007 ng/ml.
  • indomethacin produced a dose-dependant decrease in adiponectin secretion that was significant (p ⁇ 0.05) at the 2.5 and 5.0 ⁇ g/ml concentrations.
  • Acacia catechu increased adiponectin secretion relative to both the TNF ⁇ r and solvent treated 3T3-L1 adipocytes at 50 ⁇ g/ml.
  • concentrations of TNF ⁇ approaching physiologic levels Acacia catechu enhanced adiponectin secretion relative to both TNF ⁇ and the solvent controls and, surprisingly, was superior to indomethacin.
  • Ll cells, Acacia catechu, and/or apecatechin would be expected to have a positive effect on all clinical pathologies in which TNF ⁇ levels are elevated and plasma adiponectin concentrations are depressed.
  • a variety of commercial Acacia samples increase lipogenesis in the 3T3-L1 adipocyte model.
  • Acacia nilotica samples #5639, #5640 and #5659 were purchased from KDN- Vita International, Inc. (121 Stryker Lane, Units 4 & 6 Hillsborough, NJ 08844). Sample #5640 was described as bark, sample #5667 as a gum resin and sample #5669 as heartwood powder. All other samples unless indicated were described as proprietary methanol extracts of Acacia catechu bark.
  • Acacia catechu that are capable of positive modification of adipocyte physiology supporting increased insulin actions.
  • the most potent formulation was #5640 with a maximal stimulation of adiponectin stimulation achieved at 12.5 ⁇ g/ml, followed by #4909 and #5668 at 25 ⁇ g/ml and finally #5639, #5667 and #5669 at 50 ⁇ g/ml.
  • Table 12 The most potent formulation was #5640 with a maximal stimulation of adiponectin stimulation achieved at 12.5 ⁇ g/ml, followed by #4909 and #5668 at 25 ⁇ g/ml and finally #5639, #5667 and #5669 at 50 ⁇ g/ml.
  • t Adiponectin Index [Adiponectin] ⁇ est/[Adiponectin] ⁇ NF ⁇ control *Significantly increased (p ⁇ 0.05) from TNF ⁇ solvent response.
  • t Adiponectin Index [Adiponectin] ⁇ est/[Adiponectin] ⁇ NF ⁇ control *Significantly increased (p ⁇ 0.05) from TNF ⁇ solvent response.
  • Polar and non-polar solvents extract compounds from Acacia catechu capable of increasing adiponectin secretion in the TNPo/3T3-Ll adipocyte model.
  • Test Materials Large chips of Acacia catechu sample #5669 heartwood (each chip weighing between 5-10 grams) were subjected to drilling with a 5/8" metal drill bit using a standard power drill at low speed. The wood shavings were collected into a mortar, and ground into a fine powder while frozen under liquid N 2 . This powder was then sieved through a 250 micron screen to render approximately 10 g of a fine free-flowing powder.
  • Gastric fluid consisted of 2.90 g NaCl, 7.0 m concentrated, aqueous HCl, 3.2 g pepsin (800 - 2500 activity units/mg) diluted to 1000 ml with water. Final pH was 1.2.
  • the gastric fluid-heartwood suspension remained at 40° C for one hour followed by removal of the gastric fluid in vacuo. The remaining residue was then dissolved in MeOH, filtered through a 0.45 micron PTFE syringe filter and concentrated in vacuo.
  • Pioglitazone was obtained as 45 mg pioglitazone tables from a commercial source as Actos® (Takeda Pharmaceuticals, Lincolnshire, IL). The tablets were ground to a fine powder and tested at 5.0, 2.5, 1.25 and 0.625 ⁇ g pioglitazone/ml. Indomethacin was also included as an additional positive control.
  • Acacia catechu acidic and basic fractions are capable of increasing adiponectin secretion in the TNFQ/3T3-L1 adipocyte model.
  • Test Materials Acacia catechu sample #5669 was extracted according to the following procedure: Alkaline isopropyl alcohol solution, (1% (v/v) 1.5N NaOH in isopropanol,) was added to approximately 50 mg of the dry Acacia catechu heartwood powder #5669 in a 50 ml tube. The sample was then mixed briefly, sonicated for 30 minutes, and centrifuged for an hour to pellet the remaining solid material. The supernatant liquid was then filtered through 0.45 micron filter paper. The pH of the basic isopropanol used was pH 8.0, while the pH of the collected liquid was pH 7.0. A portion of the clear, filtered liquid was taken to dryness in vacuo and appeared as a white solid. This sample was termed the dried alkaline extract.
  • the remaining pelleted material was brought up in acidic isopropyl alcohol solution, (1% (v/v) 10% HCl in isopropanol,) as a red solution. This sample was mixed until the pellet material was sufficiently dispersed in the liquid and then centrifuged for 30 minutes to again pellet the remaining solid. The pale yellow supernatant fluid was passed through a 0.45 micron filter paper. The pH of the collected liquid was pH 3.0 and it was found that in raising the pH of the sample to pH 8-9 a reddish-brown precipitate was formed (dried precipitate). The precipitate was collected and dried, providing a reddish-brown solid.
  • acidic isopropyl alcohol solution 1% (v/v) 10% HCl in isopropanol
  • Interleukin-6 is a multifunctional cytokine that plays important roles in host defense, acute phase reactions, immune responses, nerve cell functions, hematopoiesis and metabolic syndrome. It is expressed by a variety of normal and transformed lymphoid and nonlymphoid cells such as adipocytes.
  • IL-6 The production of IL-6 is up-regulated by numerous signals such as mitogenic or antigenic stimulation, lipopolysaccharides, calcium ionophores, cytokines and viruses [Hibi, M., Nakajima, K., Hirano T. IL-6 cytokine family and signal transduction: a model of the cytokine system. J MoI Med. 74(1):1-12, (Jan 1996)]. Elevated serum levels have been observed in a number of pathological conditions including bacterial and viral infection, trauma, autoimmune diseases, malignancies and metabolic syndrome [Arner, P. Insulin resistance in type 2 diabetes — role of the adipokines. Curr MoI Med.;5(3):333-9, (May 2005)].
  • Test Materials Indomethacin, methylisobutylxanthine, dexamethasone, and insulin were obtained from Sigma (St. Louis, MO). The test material was a dark brown powder produced from a 50:50 (v/v) water/alcohol extract of the gum resin of Acacia catechu sample #4909 and was obtained from Bayir Chemicals (No. 68, South Cross Road, Basavanagudi, India). The extract was standardized to contain not less than 20% apecatechin. Batch No. A Cat/2304 used in this example contained 20.8% apecatechin as determined by UV analysis.
  • Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS (fetal bovine serum) characterized from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
  • Interleukin-6 Assay The IL-6 secreted into the medium was quantified using the Quantikine® Mouse IL-6 Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN). Information supplied by the manufacturer indicated that recovery of IL-6 spiked in mouse cell culture media averaged 99% with a 1 :2 dilution and the minimum detectable IL-6 concentration ranged from 1.3 to 1.8 pg/ml. All supernatant media samples were assayed undiluted. [00252] Statistical Calculations and Interpretation - All assays were preformed in duplicate. For statistical analysis, the effect of Acacia on adiponectin or IL-6 secretion was computed relative to the solvent control. Differences among the doses were determined using the student's t-test without correction for multiple comparisons; the nominal five percent probability of a type I error (one-tail) was selected.
  • f Adiponectin Index [Adiponectin] ⁇ est/[Adiponectin] ⁇ NF ⁇ control
  • ffIL-6 Index [IL-6 Tes t - IL-6 C ontrol]/[IL-6 ⁇ NF ⁇ - IL-6 C ontrol]
  • Acacia catechu sample #4909 demonstrated a dual anti-inflammatory action in the TNF ⁇ /3T3-Ll adipocyte model.
  • Components of the Acacia catechu extract increased adiponectin secretion while decreasing IL-6 secretion.
  • the overall effect of Acacia catechu was strongly anti-inflammatory relative to the TNF ⁇ controls.
  • Resistin Assay The amount of resistin secreted into the medium was quantified using the Quantikine® Mouse Resistin Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN). Information supplied by the manufacturer indicated that recovery of resistin spiked in mouse cell culture media averaged 99% with a 1:2 dilution and the minimum detectable resistin concentration ranged from 1.3 to 1.8 pg/ml. All supernatant media samples were diluted 1 :20 with dilution media supplied by the manufacturer before assay.
  • f Adiponectin Index [Adiponectin] ⁇ est /[Adiponectin] lnsu
  • jn control ttIL-6 Index [IL-6 ⁇ est]/[ ⁇ .-6
  • f ftResistin Index [Resistin ⁇ es t]/[Resistini nsu i in control]
  • Index values represent the mean ⁇ 95% confidence interval computed from residual mean square of the analysis of variance. Values greater or less than Insulin control ⁇ 95% CI are significantly different with p ⁇ 0.05.
  • Results The positive controls indomethacin and troglitazone induced
  • the positive hops phytochemical genera in this study which included isomerized alpha acids, alpha acids and beta acids as well as xanthohumols, may be expected to increase insulin sensitivity and decrease serum triglycerides in humans or other animals exhibiting signs or symptoms of insensitivity to insulin.
  • Hops phytochemicals increase adiponectin secretion in insulin-resistant 3T3-L1 adipocytes.
  • the Model The 3T3-L1 murine fibroblast model as described in Examples
  • Rho isoalpha acids 2.38 0.10
  • the concentration of test material required for stimulation of half maximal adiponectin secretion in insulin-resistant 3T3-L1 cells was similar for troglitazone, Rho isoalpha acids, tetrahydroisoalpha acid and hexahydroisoalpha acids.
  • the concentration of isoalpha acids at half maximal adiponectin secretion 0.49 ⁇ g/ml was nearly 5-fold greater.
  • Xanthohumols exhibited the lowest dose for half maximal adiponectin secretion estimated at 0.037 ⁇ g/ml.
  • 3T3-L1 cells the positive hops phytochemical genera seen in this study, isoalpha acids, Rho- isoalpha acids, tetrahydroisoalpha acids, hexahydroisoalpha acids, xanthohumols, spent hops and hexahydro colupulone, may be expected to have a positive effect on all clinical pathologies in which plasma adiponectin concentrations are depressed.
  • Hops phytochemicals exhibit anti-inflammatory activity through enhanced adiponectin secretion and inhibition of interleukin-6 secretion in insulin-resistant 3T3-L1 adipocytes.
  • Index value is mean ⁇ 95% confidence interval computed from residual mean square of the analysis of variance.
  • values ⁇ 0.7 or > 1.3 are significantly different from insulin control and for IL-6, values ⁇ 0.77 or >1.23 are significantly different from insulin control.
  • the adiponectin/IL-6 ratio was strongly positive (>2.00) for RIAA, IAA HHIA, and XN. THIAA, HHCL and spent hops exhibited positive, albeit lower, adiponectin/IL-6 ratios.
  • the adiponectin/IL-6 ratio was mixed with a strongly positive response at 2.5 and 0.625 ⁇ g/ml and no effect at 5.0 or 1.25 ⁇ g/ml.
  • Hops phytochemicals increase adiponectin secretion in TNFotreated 3T3-L1 adipocytes.
  • TNFoVmI markedly suppressed adiponectin secretion ( Figure 21).
  • the hops derivatives IAA, RIAA, HHIAA and THIAA all increased adiponectin secretion relative to the TNFoysolvent control.
  • Linear dose-response curves were observed with RIAA and HHIAA resulting in maximal inhibition at the highest concentration tested 5.0 ⁇ g/ml.
  • IAA elicited maximal secretion of adiponectin at 1.25 ⁇ g/ml, while THIAA exhibited a curvilinear response with maximal adiponectin secretion at 5.0 ⁇ g/ml.
  • hops derivatives IAA, RIAA, HHIAA and THIAA to increase adipocyte adiponectin secretion in the presence of supraphysiological concentrations of TNF ⁇ supports the usefulness of these compounds in the prevention or treatment of inflammatory conditions involving suboptimal adipocyte functioning.
  • 3T3-L1 adipocytes were treated prior to differentiation as in Example 11 for computing the lipogenic index or with TNF ⁇ as described in Example 12 for assessing the adiponectin index.
  • Acacia catechu sample #5669 as described in Example 14 was used with hops derivatives Rho-isoalpha acids and isoalpha acids as previously described.
  • Acacia catechu and the 5:1 and 10:1 combinations of Acacia:RIAA and Acacia:IAA were tested at 50, 10, 5.0 and 1.0 ⁇ g/ml.
  • RIAA and IAA were tested independently at 5.0, 2.5, 1.25 and 0.625 ⁇ g/ml.
  • Rho isoalpha acids or isoalpha acids exhibit synergistic combinations and only few antagonistic combinations with respect to increasing lipid incorporation in adipocytes and increasing adiponectin secretion from adipocytes.
  • the Model The 3T3-L1 murine adipocyte model as described in Examples
  • the non-steroidal anti-inflammatory drugs (NSAIDs) aspirin, salicylic acid, and ibuprofen were obtained from Sigma.
  • the commercial capsule formulation of celecoxib (CelebrexTM, G.D. Searle & Co. Chicago, IL) was used and cells were dosed based upon content of active ingredient.
  • Hops derivatives, ibuprofen, and celecoxib were dosed at 5.00, 2.50, 1.25 and 0.625 ⁇ g/ml.
  • Indomethacin, troglitazone, and pioglitazone were tested at 10, 5.0, 1.0 and 0.50 ⁇ g/ml.
  • Concentrations for aspirin were 100, 50.0, 25.0 and 12.5 ⁇ g/ml, while those for salicylic acid were 200, 100, 50.0 and 25.0 ⁇ g/ml.
  • IL-6 and adiponectin were assayed and data were analyzed and tabulated as previously described in Example 18 for IL-6 and Example 13 for adiponectin.
  • indomethacin, troglitazone, pioglitazone, ibuprofen and celecoxib inhibited IL-6 secretion at all concentrations tested, while RIAA, IAA, and aspirin did not significantly inhibit IL-6 at the lowest concentrations (data not shown).
  • Pioglitazone was next in order of potency with adiponectin stimulation of 12% at 1.25 ⁇ g/ml. With a 9% stimulation of adiponectin secretion at 2.50 ⁇ g/ml, indomethacin was least potent of the active test materials.
  • hops derivatives RIAA and IAA as well as ibuprofen decreased IL-6 secretion and increased adiponectin secretion at concentrations likely to be obtained in vivo.
  • the thiazolidinediones troglitazone and pioglitazone were less potent as inhibitors of IL-6 secretion, requiring higher doses than hops derivatives, but similar to hops derivatives with respect to adiponectin stimulation. No consistent relationship between anti-inflammatory activity in macrophage models and the adipocyte model was observed for the NSAIDs indomethacin, aspirin, ibuprofen and celecoxib.
  • test materials were added in concert with 100 ng LPS/ml to D5 3T3-L1 adipocytes. On the following day, supernatant media were sampled for JL-6 determination. All values were indexed to the LPS control as noted below. Concentrations presented represent dose providing the maximum inhibition of IL-6 secretion and those values less than 0.70 are significantly (p ⁇ 0.05) less than the LPS control.
  • fIL-6 Index [IL-6 Tes t - IL-6 C ⁇ ntroi]/[IL-6 L ps - IL-6 C ⁇ ntroi] *Significantly different from LPS control p ⁇ 0.05).
  • Adiponectin Index [Adiponectin] ⁇ es t/[Adiponectin]Lps control
  • the Model The 3T3-L1 murine fibroblast model as described in Examples
  • Acacia catechu sample #5669 as described in Example 14 hops derivatives Rho-isoalpha acids and xanthohumol as described in Example 20, and curcumin as provided by Metagenics (Gig Harbor, WA) and were used in these experiments.
  • Adiponectin Index [ Adiponectin] ⁇ cs /[ Adi ponectin] ⁇ NF ⁇ * control
  • Example 11 3T3-L1 adipocytes were treated prior to differentiation as in Example 11 for computing the lipogenic index.
  • Powdered CLA was obtained from Lipid Nutrition (Channahon, IL) and was described as a 1:1 mixture of the c9tl 1 and 110c 12 isomers.
  • CLA and the 5:1 combinations of CLArRIAA were tested at 50, 10, 5.0 and 1.0 ⁇ g/ml.
  • RIAA was tested at 10, 1.0 and 0.1 ⁇ g/ml for calculation of expected lipogenic index as described previously.
  • Results - RIAA synergistically increased triglyceride content in combination with CLA. Synergy was noted at all does (Table 27).
  • Hops phvtochemicals inhibit NF-kB activation in TNF ⁇ -treated 3T3-L1 adipocytes.
  • 3T3-L1 adipocytes were maintained in post-differentiation medium for an additional 40 days.
  • Standard chemicals, media and hops compounds RIAA and xanthohumol were as described in Examples 13 and 20.
  • Hops derivatives and the positive control pioglitazone were tested at concentrations of 2.5, and 5.0 ⁇ g/ml. Test materials were added 1 hour prior to and nuclear extracts were prepared three and 24 hours following treatment with TNF ⁇ .
  • ELISA - 3T3-L1 adipocytes were maintained in growth media for 40 days following differentiation.
  • Nuclear NF-kBp65 was determined using the TransAMTM NF-kB kit from Active Motif (Carlsbad, CA) was used with no modifications.
  • Jurkat nuclear extracts provided in the kit were derived from cells cultured in medium supplemented with 50 ng/ml TPA (phorbol, 12-myri state, 13 acetate) and 0.5 ⁇ M calcium ionophore A23187 for two hours at 37°C immediately prior to harvesting.
  • Protein assay Nuclear protein was quantified using the Active Motif
  • Results The TPA-treated Jurkat nuclear extract exhibited the expected increase in NF-kBp65 indicating adequate performance of kit reagents (Figure 22).
  • the PPAR.7 agonist pioglitazone did not inhibit the amount of nuclear NF-kBp65 at either three or 24 hours following TNF ⁇ treatment.
  • Nuclear translocation of NF-kBp65 was inhibited, respectively, 9.4 and 25% at 5.0 and 2.5 ⁇ g RIAA/ml at three hours post TNF ⁇ .
  • Test Chemicals and Treatment - Metformin was obtained from Sigma (St.
  • Test materials were added in dimethyl sulfoxide at Day 0 of differentiation and every two days throughout the maturation phase (Day 6/7).
  • troglitazone was added to achieve a final concentration of 4.4 ⁇ g/ml.
  • Metformin, Acacia catechu sample #5669 and the metfo ⁇ nin/Acacia combination of 1:1 (w/w) were tested at 50 ⁇ g test material/ml.
  • Differentiated 3T3-L1 cells were stained with 0.2% Oil Red O. The resulting stained oil droplets were dissolved with isopropanol and quantified by spectrophotometric analysis at 530 nm. Results were represented as a relative triglyceride content of fully differentiated cells in the solvent controls.
  • Example 11 3T3-L1 adipocytes were treated prior to differentiation as in Example 11 for computing the lipogenic index.
  • Troglitazone was obtained from Cayman Chemicals (Chicago, IL).
  • Pioglitazone was obtained as the commercial, tableted formulation (ACTOSE ® , Takeda Pharmaceuticals, Lincolnshire, IL). The tablets were crushed and the whole powder was used in the assay. All results were computed based upon active ingredient content. Hops derivatives Rho-isoalpha acids and isoalpha acids used were as described in Example 20.
  • Troglitazone in combination with RIAA and IAA was tested at 4.0 ⁇ g/ml, while the more potent pioglitazone was tested in 1 :1 combinations with RIAA and IAA at 2.5 ⁇ g/ml. All materials were also tested independently at 4.0 and 2.5 ⁇ g/ml for calculation of expected lipogenic index as described in Example 34.
  • Hops derivatives Rho-isoalpha acids and isoalpha acids could synergistically increase the insulin sensitizing effects of thiazolidinediones resulting in potential clinical benefits of dose-reduction or increased numbers of patients responding favorably.
  • Example 20 Rho-isoalpha acids were as described in Example 20. Metformin at 50, 10, 5.0 or 1.0 ⁇ g/ml without or with 1 ⁇ g RIAA/ml was added in concert with 10 ng TNFo/ml to D5 3T3-L1 adipocytes. Culture supernatant media were assayed for IL-6 on Day 6 as detailed in Example 11. An estimate of the expected effect of the metformin :RIAA mixtures on IL-6 inhibition was made as previously described.
  • Results - TNFa provided a six-fold increase in IL-6 secretion in D5 adipocytes.
  • Troglitazone at 1 ⁇ g/ml inhibited IL-6 secretion 34 percent relative to the controls, while 1 ⁇ g RIAA inhibited DL-6 secretion 24 percent relative to the controls (Table 29).
  • Metformin in combination with 1 ⁇ g RIAA/ml demonstrated synergy at the 50 ⁇ g/ml concentration and strong synergy at the 1 ⁇ g/ml concentration.
  • 1 ⁇ g RIAA provided an additional 10 percent inhibition in the mixture; while at 1 ⁇ g metformin, 1 ⁇ g RIAA increased IL-6 inhibition by 35 percent. Antagonism and no effect, respectively, were seen of the metformin:RIAA combinations at the two mid-doses.
  • Combinations of metformin and Rho-isoalpha acids function synergistically at both high and low concentrations to reduce IL-6 secretion from TNFor-treated 3T3-L1 adipocytes.
  • test materials were added in concert with 10 ng TNTWmI to D5 3T3-L1 adipocytes at the stated concentrations. On the following day, supernatant media were sampled for IL-6 determination. All values were indexed to the TNF ⁇ control.
  • fIL-6 Index [IL-6 Tes t - IL-6 Con troi]/[IL-6 ⁇ NF ⁇ - IL-6controi]
  • test compounds of the present invention were examined in the RL 95-2 endometrial cancer cell model (an over expressor of AKT kinase), and in the HT-29 (constitutively expressing COX-2) and SW480 (constitutively expressing activated AKT kinase) colon cancer cell models. Briefly, the target cells were plated into 96 well tissue culture plates and allowed to grow until subconfluent. The cells were then treated for 72 hours with various amounts of the test compounds as described in Example 4 and relative cell proliferation determined by the CyQuant (Invitrogen, Carlsbad, CA) commercial fluorescence assay.
  • Results - RL 95-2 cells were treated for 72 hours with 10 ⁇ g/ml of MgDHIAA
  • This mouse strain is the result of hybridization between the KK strain, developed in the 1940s as a model of diabetes and a strain of A y /a genotype.
  • the observed phenotype is the result of polygenic mutations that have yet to be fully characterized but at least four quantitative trait loci have been identified. One of these is linked to a missense mutation in the leptin receptor. Despite this mutation the receptor remains functional although it may not be fully efficient.
  • the KK strain develops diabetes associated with insensitivity to insulin and glucose intolerance but not overt hyperglycemia.
  • the A y mutation is a 170kb deletion of the RaIy gene that is located 5' to the agouti locus and places the control for agouti under the RaIy promoter. Homozygote animals die before implantation.
  • Test Materials Acacia nilotica sample #5659 as described in Example 14 and hops derivatives Rho-isoalpha acids, isoalpha acids and xanthohumols as described in Example 20 were used.
  • the Acacia nilotica, RIAA and IAA were administered at 100 mg/kg/day, while XN was dosed at 20 mg/kg. Additionally, 5:1 and 10:1 combinations of Acacia nilotica with RIAA, IAA and XN were formulated and dosed at 100 mg/kg/day.
  • Serum was collected from the retroorbital sinus before the initial dose and ninety minutes after the third and final dose.
  • Non-fasting serum glucose was determined enzymatically by the mutarotase/glucose oxidase method and serum insulin was determined by a mouse specific ELISA (enzyme linked immunosorbent assay).
  • mice The Model - Male, C57BLKS/J m + /m + Lepr* (db/db) mice were used to assess the potential of the test materials to reduce fasting serum glucose or insulin concentrations. This strain of mice is resistant to leptin by virtue of the absence of a functioning leptin receptor. Elevations of plasma insulin begin at 10 to 14 days and of blood sugar at 4 to 8 weeks. At the time of testing (9 weeks) the animals were markedly obese 50 ⁇ 5 g and exhibited evidence of islet hypertrophy.
  • Test Materials The positive controls metformin and rosiglitazone were dosed, respectively, at 300 mg/kg-day and 1.0 mg/kg-day for each of three consecutive days. Acacia nilotica sample #5659, hops derivatives and their combinations were dosed as described previously.
  • Serum was collected from the retroorbital sinus before the initial dose and ninety minutes after the third and final dose.
  • Non-fasting serum glucose was determined e ⁇ zymatically by the mutarotase/glucose oxidase method and serum insulin was determined by a mouse specific ELISA.
  • Results The positive controls metformin and rosiglitazone decreased both serum glucose and insulin concentrations relative to the controls (Table 31). Only RIAA and XN demonstrated acceptable results as single test materials. RIAA reduced serum insulin, while XN produced a reduction in serum glucose with no effect on insulin. Acacia:RIAA [5:1] was the most effective agent tested for reducing serum insulin concentrations, providing a 21 percent reduction in serum insulin levels versus a 17 percent reduction in insulin concentrations by the biguanide metformin and a 15 percent decrease by the thiazolidinedione rosiglitazone.
  • Rho-isoalpha acids and Acacia catechu appeared synergistic in the db/db murine diabetes model.
  • the positive responses exhibited by Rho-isoalpha acids, xanthohumols and the AcaciarRIAA [5:1] formulation in two independent animal models of diabetes and three in vitro models supports their potential usefulness in clinical situations requiring a reduction in serum glucose or enhance insulin sensitivity.
  • mice The Model - Male, C57BLKS/J m+/m+ heprdb (db/db) mice were used to assess the potential of the test materials to reduce fasting serum glucose or insulin concentrations. This strain of mice is resistant to leptin by virtue of the absence of a functioning leptin receptor. Elevations of plasma insulin begin at 10 to 14 days and of blood sugar at 4 to 8 weeks. At the time of testing (9 weeks) the animals were markedly obese 50 ⁇ 5 g and exhibited evidence of islet hypertrophy.
  • [00335J Test Materials The positive controls metformin and rosiglitazone were dosed, respectively, at 300 mg/kg-day and 1.0 mg/kg-day for each of five consecutive days.
  • Serum was collected from the retroorbital sinus before the initial dose and ninety minutes after the fifth and final dose.
  • Non-fasting serum glucose was determined enzymatically by the mutarotase/glucose oxidase method and serum insulin was determined by a mouse specific ELISA.
  • THIAA in reducing inflamation and arthritic symptomology in a rheumatoid arthritis model, such inflammation and symptoms being known to mediated, in part, by a number of protein kinases.
  • mice The Model - Female DBA/J mice (10/group) were housed under standard conditions of light and darkness and allow diet ad libitum. The mice were injected intradermally on day 0 with a mixture containing 100 ⁇ g of type II collagen and 100 ⁇ g of Mycobacterium tuberculosis in squalene. A booster injection was repeated on day 21. Mice were examined on days 22 — 27 for arthritic signs with nonresponding mice removed from the study. Mice were treated daily by gavage with test compounds for 14 days beginning on day 28 and ending on day 42.
  • Test compounds as used in this example were RIAA (MgRho) at 10 mg/kg (lo), 50 mg/kg (med), or 250 mg/kg (hi); THIAA at 10 mg/kg (lo), 50 mg/kg (med), or 250 mg/kg (hi); celecoxib at 20 mg/kg; and prednisilone at 10 mg/kg.
  • mice were euthanized and one limb, was removed and preserved in buffered formalin. After the analysis of the arthritic index was found to be encouraging, two animals were selected at random from each treatment group for histological analysis by H&E staining. Soft tissue, joint and bone changes were monitored on a four point scale with a score of 4 indicating severe damage.
  • Cytokine analysis - Serum was collected from the mice at the termination of the experiment for cytokine analysis.
  • the volume of sample being low ⁇ 0.2- 0.3 ml/mouse
  • samples from the ten mice were randomly allocated into two pools of five animals each. This was done so to permit repeat analyses; each analysis was performed a minimum of two times.
  • TNF-ct and IL-6 were analyzed using mouse specific reagents (R&D Systems, Minneapolis, W
  • Figure 29 Significant reductions (p ⁇ 0.05,two tail t-test) were observed for prednisolone at 10 mg/kg (days 30 - 42), celecoxib at 20 mg/kg (days 32 - 42), RIAA at 250 mg/kg (days 34 - 42) and RIAA at 50 mg/kg (days 38 - 40), demonstrating antiarthritc efficacy for RIAA at 50 or 250 mg/kg .
  • Figure 30 displays the effects of THIAA on the arthritic index.
  • RIAA Acacia (1:5) effects on metabolic syndrome in humans.
  • subjects had to meet 3 of the following 5 criteria: (i) waist circumference > 35" (women) and > 40" (men); (ii) TG >150 mg/dL; (iii) HDL ⁇ 50 mg/dL (women), and ⁇ 40 mg/dL (men); (iv) blood pressure > 130/85 or diagnosed hypertension on medication; and (v) fasting glucose ⁇ IOO mg/dL.
  • the homeostatic model assessment (HOMA) score is a published measure of insulin resistance.
  • the change in HOMA score for all subjects is shown in Figure 35. Due to the variability seen in metabolic syndrome subjects' insulin and glucose values, a subgroup of only those subjects with fasting insulin > 15 mcIU/mL was also assessed.
  • the HOMA score for this subgroup is shown in Table 33, and indicates that a significant decrease was observed for the RIAA/Acacia group as compared to the placebo group.
  • HOMA score was calculated from fasting insulin and glucose by published methods [(insulin (mcrU/mL)*glucose (mg/dL))/405].
  • Elevation in triglycerides is also an important suggestive indicator of metabolic syndrome.
  • Table 34 and Figure 36 indicate that RIAA/Acacia supplementation resulted in a significant decrease in TG after 8 weeks as compared with placebo (p ⁇ 0.05).
  • the TG/HDL-C ratio was also shown to decrease substantially for the RIAA/Acacia group (from 6.40 to 5.28), while no decrease was noted in the placebo group (from 5.81 to 5.92).
  • OBSERVED percent decrease in viable cells relative to the DMSO solvent control was computed.
  • Graphed OBSERVED values are means of eight observations ⁇ 95% confidence intervals. Synergy was inferred when the ESTIMATED percent decrease fell below the 95% confidence interval of the corresponding OBSERVED fraction.
  • Figures 42 and 43 graphically present a comparison between the observed and expected inhibitory effects of RIAA (Fig. 42) or THIAA (Fig. 43) on cancer cell proliferation. These results indicate that the compounds tested in combination with celecoxib inhibited cancer cell proliferation to an extent greater than mathematically predicted in most instances.
  • Figure 44 graphically displays the detection of THIAA in the serum over time following ingestion of 940 mg of THIAA.
  • Figure 45 demonstrates that after 225 minutes following ingestion, THIAA was detected in the serum at levels comparabe to those THIAA levels tested in vitro.
  • Figure 46 depicts the metabolism of THIAA by CYP2C9*1.

Abstract

Compounds and methods for protein kinase modulation for cancer treatment are disclosed. The compounds and methods disclosed are based on hexahydro-isoalpha acids, commonly found in hops.

Description

HEXAHYDRO-ISOALPHA ACID BASED PROTEIN KINASE MODULATION
CANCER TREATMENT
CROSS-REFERENCE TO RELATED APPLICATIONS
[001] This patent application claims priority to U.S. provisional applications Ser.
No. 60/815,064 filed on June 20, 2006.
BACKGROUND OF THE INVENTION
Field of the Invention
[002] The present invention relates generally to methods and compositions that can be used to treat or inhibit cancers susceptible to protein kinase modulation. More specifically, the invention relates to methods and compositions which utilize compounds or derivatives commonly isolated either from hops or from members of the plant genus Acacia, or combinations thereof.
Description of the Related Art
[003] Signal transduction provides an overarching regulatory mechanism important to maintaining normal homeostasis or, if perturbed, acting as a causative or contributing mechanism associated with numerous disease pathologies and conditions. At the cellular level, signal transduction refers to the movement of a signal or signaling moiety from outside of the cell to the cell interior. The signal, upon reaching its receptor target, may initiate ligand-receptor interactions requisite to many cellular events, some of which may further act as a subsequent signal. Such interactions serve to not only as a series cascade but moreover an intricate interacting network or web of signal events capable of providing fine-tuned control of homeostatic processes. This network however can become dysregulated, thereby resulting in an alteration in cellular activity and changes in the program of genes expressed within the responding cell. See, for example, Figure 1 which displays a simplified version of the interacting kinase web regulating insulin sensitivity and resistance. fOO4] Signal transducing receptors are generally classified into three classes. The first class of receptors are receptors that penetrate the plasma membrane and have some intrinsic enzymatic activity. Representative receptors that have intrinsic enzymatic activities include those that are tyrosine kinases (e.g. PDGF, insulin, EGF and FGF receptors), tyrosine phosphatases (e.g. CD45 [cluster determinants] protein of T cells and macrophages), guanylate cyclases (e.g. natriuretic peptide receptors) and serine/threonine kinases (e.g. activin and TGF-/3 receptors). Receptors with intrinsic tyrosine kinase activity are capable of autophosphorylation as well as phosphorylation of other substrates.
[005] Receptors of the second class are those that are coupled, inside the cell, to
GTP-binding and hydrolyzing proteins (termed G-proteins). Receptors of this class which interact with G-proteins have a structure that is characterized by 7 transmembrane spanning domains. These receptors are termed serpentine receptors. Examples of this class are the adrenergic receptors, odorant receptors, and certain hormone receptors (e.g. glucagon, angiotensin, vasopressin and bradykinin).
[006] The third class of receptors may be described as receptors that are found intracellularly and, upon ligand binding, migrate to the nucleus where the ligand-receptor complex directly affects gene transcription.
[007] The proteins which encode for receptor tyrosine kinases (RTK) contain four major domains, those being: a) a transmembrane domain, b) an extracellular ligand binding domain, c) an intracellular regulatory domain, and d) an intracellular tyrosine kinase domain. The amino acid sequences of RTKs are highly conserved with those of cAMP-dependent protein kinase (within the ATP and substrate binding regions). RTK proteins are classified into families based upon structural features in their extracellular portions which include the cysteine rich domains, irnmunoglobulin-like domains, cadherin domains, leucine-rich domains, Kringle domains, acidic domains, fibronectin type III repeats, discoidin I-like domains, and EGF-like domains. Based upon the presence of these various extracellular domains the RTKs have been sub-divided into at least 14 different families.
[008] Many receptors that have intrinsic tyrosine kinase activity upon phosphorylation interact with other proteins of the signaling cascade. These other proteins contain a domain of amino acid sequences that are homologous to a domain first identified in the c-Src proto-oncogene. These domains are termed SH2 domains. [009] The interactions of SH2 domain containing proteins with RTKs or receptor associated tyrosine kinases leads to tyrosine phosphorylation of the SH2 containing proteins. The resultant phosphorylation produces an alteration (either positively or negatively) in that activity. Several SH2 containing proteins that have intrinsic enzymatic activity include phospholipase C-γ (PLC-7), the proto-oncogene c-Ras associated GTPase activating protein (rasGAP), phosphatidylinositol-3 -kinase (PI-3K), protein tyrosine phosphatase- 1C (PTPlC), as well as members of the Src family of protein tyrosine kinases (PTKs).
[0010] Non-receptor protein tyrosine kinases (PTK) by and large couple to cellular receptors that lack enzymatic activity themselves. An example of receptor-signaling through protein interaction involves the insulin receptor (IR). This receptor has intrinsic tyrosine kinase activity but does not directly interact, following autophosphorylation, with enzymatically active proteins containing SH2 domains (e.g. PI-3K or PLC-γ). Instead, the principal IR substrate is a protein termed IRS-I.
[0011] The receptors for the TGF-β superfamily represent the prototypical receptor serine/threonine kinase (RSTK). Multifunctional proteins of the TGF- β superfamily include the activins, inhibins and the bone morphogenetic proteins (BMPs). These proteins can induce and/or inhibit cellular proliferation or differentiation and regulate migration and adhesion of various cell types. One major effect of TGF-/? is a regulation of progression through the cell cycle. Additionally, one nuclear protein involved in the responses of cells to TGF-/3 is c-Myc, which directly affects the expression of genes harboring Myc-binding elements. PKA, PKC, and MAP kinases represent three major classes of non-receptor serine/threonine kinases.
[0012] The relationship between kinase activity and disease 'States is currently being investigated in many laboratories. Such relationships may be either causative of the disease itself or intimately related to the expression and progression of disease associated symptomology. Rheumatoid arthritis, an autoimmune disease, provides one example where the relationship between kinases and the disease are currently being investigated.
[0013] Autoimmune diseases result from a dysfunction of the immune system in which the body produces autoantibodies which attack its own organs, tissues and cells — a process mediated via protein phosphorylation. [0014] Over 80 clinically distinct autoimmune diseases have been identified and collectively afflict approximately 24 million people in the US. Autoimmune diseases can affect any tissue or organ of the body. Because of this variability, they can cause a wide range of symptoms and organ injuries, depending upon the site of autoimmune attack. Although treatments exist for many autoimmune diseases, there are no definitive cures for any of them. Treatments to reduce the severity often have adverse side effects.
[0015] Rheumatoid arthritis (RA) is the most prevalent and best studied of the autoimmune diseases and afflicts about 1% of the population worldwide, and for unknown reasons, like other autoimmune diseases, is increasing. RA is characterized by chronic synovial inflammation resulting in progressive bone and cartilage destruction of the joints. Cytokines, chemokines, and prostaglandins are key mediators of inflammation and can be found in abundance both in the joint and blood of patients with active disease. For example, PGE2 is abundantly present in the synovial fluid of RA patients. Increased PGE2 levels are mediated by the induction of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) at inflamed sites. [See, for example van der Kraan PM and van den Berg WB. Anabolic and destructive mediators in osteoarthritis. Curr Opin Clin Nutr Metab Care,3:205- 211, 2000; Choy EHS and Panayi GS. Cytokine pathways and joint inflammation in rheumatoid arthritis. N Eng J Med. 344:907-916, 2001; and Wong BR, et al. Targeting Syk as a treatment for allergic and autoimmune disorders. Expert Opin Investig Drugs 13:743-762, 2004.]
[0016] The etiology and pathogenesis of RA in humans is still poorly understood, but is viewed to progress in three phases. The initiation phase where dendritic cells present self antigens to autoreactive T cells. The T cells activate autoreactive B cells via cytokines resulting in the production of autoantibodies, which in turn form immune complexes in joints. In the effector phase, the immune complexes bind Fcf receptors on macrophages and mast cells, resulting in release of cytokines and chemokines, inflammation and pain. In the final phase, cytokines and chemokines activate and recruit synovial fibroblasts, osteoclasts and polymorphonuclear neutrophils that release proteases, acids, and ROS such as O2-, resulting in irreversible cartilage and bone destruction.
[0017] In the collagen-induced RA animal model, the participation of T and B cells is required to initiate the disease. B cell activation signals through spleen tyrosine kinase (Syk) and phosphoinositide 3-kinase (PI3K) following antigen receptor triggering [Ward SG,- Finan P. Isoform-specific phosphoinositide 3-kinase inhibitors as therapeutic agents. Curr Opin Pharmacol. Aug;3(4):426-34, (2003)]. After the engagement of antigen receptors on B cells, Syk is phosphorylated on three tyrosines. Syk is a 72-kDa protein-tyrosine kinase that plays a central role in coupling immune recognition receptors to multiple downstream signaling pathways. This function is a property of both its catalytic activity and its ability to participate in interactions with effector proteins containing SH2 domains. Phosphorylation of Tyr-317, - 342, and -346 create docking sites for multiple SH2 domain containing proteins. [Hutchcroft, J. E., Harrison, M. L. & Geahlen, R. L. (1992). Association of the 72-kDa protein-tyrosine kinase Ptk72 with the B-cell antigen receptor. J. Biol. Chem. 267: 8613-8619, (1992) and Yamada, T., Taniguchi, T., Yang, C, Yasue, S., Saito, H. & Yamamura, H. Association with B-cell antigen cell antigen receptor with protein-tyrosine kinase-P72(Syk) and activation by engagement of membrane IgM. Eur. J. Biochem. 213: 455-459,(1993)].
[0018] Syk has been shown to be required for the activation of PI3K in response to a variety of signals including engagement of the B cell antigen receptor (BCR) and macrophage or neutrophil Fc receptors. [See Crowley, M. T., et al,. J. Exp. Med. 186: 1027- 1039, (1997); Raeder, E. M., et al., J. Immunol. 163, 6785-6793, (1999); and Jiang, K., et al., Blood 101, 236-244, (2003)]. In B cells, the BCR-stimulated activation of PI3K can be accomplished through the phosphorylation of adaptor proteins such as BCAP, CDl 9, or Gabl, which creates binding sites for the p85 regulatory subunit of PI3K. Signals transmitted by many IgG receptors require the activities of both Syk and PI3K and their recruitment to the site of the clustered receptor. In neutrophils and monocytes, a direct association of PI3K ' with phosphorylated immunoreceptor tyrosine based activation motif sequences on FcgRIIA was proposed as a mechanism for the recruitment of PI3K to the receptor. And recently a direct molecular interaction between Syk and PI3K has been reported [Moon KD, et al., Molecular Basis for a Direct Interaction between the Syk Protein-tyrosine Kinase and Phosphoinositide 3-Kinase. J. Biol. Chem. 280, No. 2, Issue of January 14, pp. 1543—1551, (2005)].
[0019] Much research has shown that inhibitors of COX-2 activity result in decreased production of PGE2 and are effective in pain relief for patients with chronic arthritic conditions such as RA. However, concern has been raised over the adverse effects of agents that inhibit COX enzyme activity since both COX-I and COX-2 are involved in important maintenance functions in tissues such as the gastrointestinal and cardiovascular systems. Therefore, designing a safe, long term treatment approach for pain relief in these patients is necessary. Since inducers of COX-2 and iNOS synthesis signal through the Syk, PBK, p38, ERK1/2, and NF-kB dependent pathways, inhibitors of these pathways may be therapeutic in autoimmune conditions and in particular in the inflamed and degenerating joints of RA patients.
[0020] The hops derivative Rho isoalpha acid (RIAA) was found in a screen for inhibition of PGE2 in a RAW 264.7 mouse macrophages model of inflammation. In the present study, we investigated whether RIAA is a direct COX enzyme inhibitor and/or whether it inhibits the induction of COX-2 and iNOS. Our finding that RIAA does not directly inhibit COX enzyme activity, but instead inhibits NF-kB driven enzyme induction lead us to investigate whether RIAA is a kinase inhibitor. Our finding that RIAA inhibits both Syk and PI3K lead us to test its efficacy in a pilot study in patients suffering from various autoimmunine diseases.
[0021] Other kinases currently being investigated for their association with disease symptomology include Aurora, FGFB, MSK, RSE, and SYK.
[0022] Aurora - Important regulators of cell division, the Aurora family of serine/threonine kinases includes Aurora A, B and C. Aurora A and B kinases have been identified to have direct but distinct roles in mitosis. Over-expression of these three iso forms have been linked to a diverse range of human tumor types, including leukemia, colorectal, breast, prostate, pancreatic, melanoma and cervical cancers.
[0023] Fibroblast growth factor receptor (FGFR) is a receptor tyrosine kinase.
Mutations in this receptor can result in constitutive activation through receptor dimerization, kinase activation, and increased affinity for FGF. FGFR has been implicated in achondroplasia, angiogenesis, and congenital diseases.
[0024] MSK (mitogen- and stress-activated protein kinase) 1 and MSK2 are kinases activated downstream of either the ERK (extracellular-signal-regulated kinase) 1/2 or p38 MAPK (mitogen-activated protein kinase) pathways in vivo and are required for the phosphorylation of CREB (cAMP response element-binding protein) and histone H3.
[0025] Rse is mostly highly expressed in the brain. Rse, also known as Bit, BYK,
Dtk, Etk3, Sky, Tif, or sea-related receptor tyrosine kinase, is a receptor tyrosine kinase whose primary role is to protect neurons from apoptosis. Rse, AxI, and Mer belong to a newly identified family of cell adhesion molecule-related receptor tyrosine kinases. GAS6 is a Hgand for the tyrosine kinase receptors Rse, AxI, and Mer. GAS6 functions as a physiologic anti-inflammatory agent produced by resting EC and depleted when proinflammatory stimuli turn on the pro-adhesive machinery of EC.
[0026] Glycogen synthase kinase-3 (GSK-3), present in two isoforms, has been identified as an enzyme involved in the control of glycogen metabolism, and may act as a regulator of cell proliferation and cell death. Unlike many serine-threonine protein kinases, GSK-3 is constitutively active and becomes inhibited in response to insulin or growth factors. Its role in the insulin stimulation of muscle glycogen synthesis makes it an attractive target for therapeutic intervention in diabetes and metabolic syndrome.
{0027] GSK-3 dysregulation has been shown to be a focal point in the development of insulin resistance. Inhibition of GSK3 improves insulin resistance not only by an increase of glucose disposal rate but also by inhibition of gluconeogenic genes such as phosphoenolpyruvate carboxykinase and glucose-6-phosphatase in hepatocytes. Furthermore, selective GSK3 inhibitors potentiate insulin-dependent activation of glucose transport and utilization in muscle in vitro and in vivo. GSK.3 also directly phosphorylates serine/threonine residues of insulin receptor substrate- 1, which leads to impairment of insulin signaling. GSK3 plays an important role in the insulin signaling pathway and it phosphorylates and inhibits glycogen synthase in the absence of insulin [Parker, P. J., Caudwell, F. B., and Cohen, P. (1983) Eur. J. Biochem. 130:227-234]. Increasing evidence supports a negative role of GSK-3 in the regulation of skeletal muscle glucose transport activity. For example, acute treatment of insulin-resistant rodents with selective GSK-3 inhibitors improves whole-body insulin sensitivity and insulin action on muscle glucose transport. Chronic treatment of insulin-resistant, pre-diabetic obese Zucker rats with a specific GSK-3 inhibitor enhances oral glucose tolerance and whole-body insulin sensitivity, and is associated with an amelioration of dyslipidemia and an improvement in IRS-I- dependent insulin signaling in skeletal muscle. These results provide evidence that selective targeting of GSK-3 in muscle may be an effective intervention for the treatment of obesity- associated insulin resistance. [0028] Syk is a non-receptor tyrosine kinase related to ZAP-70 involved in signaling from the B-cell receptor and the IgE receptor. Syk binds to ITAM motifs within these receptors, and initiates signaling through the Ras, PI 3-kinase, and PLCg signaling pathways. Syk plays a critical role in intracellular signaling and thus is an important target for inflammatory diseases and respiratory disorders.
[0029] Therefore, it would be useful to identify methods and compositions that would modulate the expression or activity of single or multiple selected kinases. The realization of the complexity of the relationship and interaction among and between the various protein kinases and kinase pathways reinforces the pressing need for developing pharmaceutical agents capable of acting as protein kinase modulators, regulators or inhibitors that have beneficial activity on multiple kinases or multiple kinase pathways. A single agent approach that specifically targets one kinase or one kinase pathway may be inadequate to treat very complex diseases, conditions and disorders, such as, for example, diabetes and metabolic syndrome. Modulating the activity of multiple kinases may additionally generate synergistic therapeutic effects not obtainable through single kinase modulation.
[0030] Such modulation and use may require continual use for chronic conditions or intermittent use, as needed for example in inflammation, either as a condition unto itself or as an integral component of many diseases and conditions. Additionally, compositions that act as modulators of kinase can affect a wide variety of disorders in a mammalian body. The instant invention describes compounds and extracts derived from hops or Acacia which may be used to regulate kinase activity, thereby providing a means to treat numerous disease related symptoms with a concomitant increase in the quality of life.
SUMMARY OF THE INVENTION
[0031] The present invention relates generally to methods and compositions that can be used to treat or inhibit cancers susceptible to protein kinase modulation. More specifically, the invention relates to methods and compositions which utilize compounds or derivatives commonly isolated either from hops or from members of the plant genus Acacia, or combinations thereof. [0032) A first embodiment of the invention describes methods to treat a cancer responsive to protein kinase modulation in a mammal in need. The method comprises administering to the mammal a therapeutically effective amount of a hexahydro-isoalpha acid.
[0033] A second embodiment of the invention describes compositions to treat a cancer responsive to protein kinase modulation in a mammal in need where the composition comprises a therapeutically effective amount of a hexahydro-isoalpha acid where the therapeutically effective amount modulates a cancer associated protein kinase.
BRIEF DESCRIPTION OF THE DRAWINGS
10034] Figure 1 graphically depicts a portion of the kinase network regulating insulin, sensitivity and resistance.
10035] Figure 2 graphically depicts the inhibition of five selected kinases by
MgRIAA (mgRho).
[0036] Figure 3 graphically depicts the inhibition of PI3K isoforms by five hops components and a Acacia nilotica extract.
[0037] Figure 4 depicts RIAA [panel A] and IAA [panel B] dose-related inhibition of
PGE2 biosynthesis when added before LPS stimulation of COX-2 expression (white bars) or following overnight LPS-stimulation prior to the addition of test material (grey bars).
[0038] Figure 5 provides a graphic representation of direct enzymatic inhibition of celecoxib [panel A] and MgRIAA [panel B] on LPS induced COX-2 mediated PGE2 production analyzed in RAW 264.7 cells. PGE2 was measured and expressed in pg/ml. The error bars represent the standard deviation (n = 8).
[0039] Figure 6 provides Western blot detection of COX-2 protein expression. RAW
264.7 cells were stimulated with LPS for the indicated times, after which total cell extract was visualized by western blot for COX-2 and GAPDH expression [panel A]. Densitometry of the COX-2 and GAPDH bands was performed. The graph [panel B] represents the ratio of COX-2 to GAPDH. [0040] Figure 7 provides Western blot detection of iNOS protein expression. RAW
264.7 cells were stimulated with LPS for the indicated times, after which total cell extract was visualized by western blot for iNOS and GAPDH expression [panel A]. Densitometry of the iNOS and GAPDH bands was performed. The graph [panel B] represents the ratio of iNOS to GAPDH.
[0041] Figure 8 provides a representative schematic of the TransAM NF-κB kit utilizing a 96-well format. The oligonucleotide bound to the plate contains the consensus binding site for NF-κB. The primary antibody detected the p50 subunit of NF-ΛB.
[0042] Figure 9 provides representative binding activity of NF-κB as determined by the TransAM NF-κB kit. The percent of DNA binding was calculated relative to the LPS control (100%). The error bars represent the standard deviation (n = 2). RAW 264.7 cells were treated with test compounds and LPS for 4 hr as described in the Examples section.
[0043] Figure 10 is a schematic of a representative testing procedure for assessing the lipogenic effect of an Acacia sample #4909 extract on developing and mature adipocytes. The 3T3-L1 murine fibroblast model was used to study the potential effects of the test compounds on adipocyte adipogenesis.
[0044] Figure 11 is a graphic representation depicting the nonpolar lipid content of
3T3-L1 adipocytes treated with an Acacia sample #4909 extract or the positive controls indomethacin and troglitazone relative to the solvent control. Error bars represent the 95% confidence limits (one-tail).
[0045] Figure 12 is a schematic of a representative testing procedure for assessing the effect of a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia sample #4909 on the secretion of adiponectin from insulin-resistant 3T3-L1 adipocytes.
[0046] Figure 13 is a representative bar graph depicting maximum adiponectin secretion by insulin-resistant 3T3-L1 cells in 24 hr elicited by three doses of troglitazone and four doses of a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia sample #4909. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals. {0047] Figure 14 is a schematic of a representative testing protocol for assessing the effect of a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia sample #4909 on the secretion of adiponectin from 3T3-L1 adipocytes treated with test material plus 10, 2 or 0.5 πg TNFα/ml.
[0048] Figure 15 depicts representative bar graphs representing adiponectin secretion by TNFα treated mature 3T3-L1 cells elicited by indomethacin or an Acacia sample #4909 extract. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals. *Significantly different from TNFo- alone treatment (p<0.05).
[0049] Figure 16 graphically illustrates the relative increase in triglyceride content in insulin resistant 3T3-L1 adipocytes by various compositions of Acacia catechu and A. nilotica from different commercial sources. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals.
[0050] Figure 17 graphically depicts a representation of the maximum relative adiponectin secretion elicited by various extracts of Acacia catechu. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals.
[0051] Figure 18 graphically depicts the lipid content (relative to the solvent control) of 3T3-L1 adipocytes treated with hops compounds or the positive controls indomethacin and troglitazone. The 3T3-L1 murine fibroblast model was used to study the potential effects of the test compounds on adipocyte adipogenesis. Results are represented as relative nonpolar lipid content of control cells; error bars represent the 95% confidence interval.
[0052] Figure 19 is a representative bar graph of maximum adiponectin secretion by insulin-resistant 3T3-L1 cells in 24 hr elicited by the test material over four doses. Values presented are as a percent relative to the solvent control; error bars represent 95% confidence intervals. IAA = isoalpha acids, RIAA = Rho isoalpha acids, HHIA = hexahydroisoalpha acids, and THIAA = tetrahydroisoalpha acids.
[0053] Figure 20 depicts the Hofstee plots for Rho isoalpha acids, isoalpha acids, tetrahydroisoalpha acids, hexahydroisoalpha acids, xanthohumols, spent hops, hexahydrocolupulone and the positive control troglitazone. Maximum adiponectin secretion relative to the solvent control was estimated from the y-intercept, while the concentration of test material necessary for half maximal adiponectin secretion was computed from the negative value of the slope.
[0054] Figure 21 displays two bar graphs representing relative adiponectin secretion by TNFα-treated, mature 3T3-L1 cells elicited by isoalpha acids and Rho isoalpha acids [panel A], and hexahydro isoalpha acids and tetrahydro isoalpha acids [panel B]. Values presented are percent relative to the solvent control; error bars represent 95% confidence intervals. *Significantly different from TNFo; only treatment (p<0.05).
[0055] Figure 22 depicts NF-kB nuclear translocation in insulin-resistant 3T3-L1 adipocytes [panel A] three and [panel B] 24 hr following addition of 10 ng TNFo/ml. Pioglitazone, RIAA and xanthohumols were added at 5.0 (black bars) and 2.5 (stripped bars) μg/ml. Jurkat nuclear extracts from cells cultured in medium supplemented with 50 ng/ml TPA (phorbol, 12-myristate, 13 acetate) and 0.5 μM calcium ionophore A23187 (CI) for two hours at 37°C immediately prior to harvesting.
[0056] Figure 23 graphically describes the relative triglyceride content of insulin resistant 3T3-L1 cells treated with solvent, metformin, an Acacia sample #5659 aqueous extract or a 1:1 combination of metfoτmin/Acacia catechu extract. Results are represented as a relative triglyceride content of fully differentiated cells in the solvent controls.
[0057] Figure 24 graphically depicts the effects of 10 μg/ml of solvent control
(DMSO), RIAA, isoalpha acid (IAA), tetrahydroisoalpha acid (THIAA), a 1:1 mixture of THIAA and hexahydroisoalpha acid (HHIAA), xanthohumol (XN), LY 249002 (LY), ethanol (ETOH), alpha acid (ALPHA), and beta acid (BETA) on cell proliferation in the RL 95-2 endometrial cell line.
[0058] Figure 25 graphically depicts the effects of various concentrations of THIAA or reduced isoalpha acids (RIAA) on cell proliferation in the HT-29 cell line.
[0059] Figure 26 graphically depicts the effects of various concentrations of THIAA or reduced isoalpha acids (RIAA) on cell proliferation in the SW480 cell line.
[0060] Figure 27 graphically depicts the dose responses of various combinations of reduced isoalpha acids (RIAA) and Acacia for reducing serum glucose [panel A] and serum insulin [panel B] in the db/db mouse model. [0061] Figure 28 graphically depicts the reduction in serum glucose [panel A] and serum insulin [panel B] in the db/db mouse model produced by a 5:1 combination of RIAA:Acacia as compared to the pharmaceutical anti-diabetic compounds roziglitazone and metformin.
[0062] Figure 29 graphically depicts the effects of reduced isoalpha acids (RIAA) on the arthritic index in a murine model of rheumatoid arthritis.
[0063J Figure 30 graphically depicts the effects of THIAA on the arthritic index in a murine model of rheumatoid arthritis.
[0064] Figure 31 graphically summarizes the effects of RIAA and THIAA on collagen induced joint damage.
[0065] Figure 32 graphically summarizes the effects of RIAA and THIAA on IL-6 levels in a collagen induced arthritis animal model.
[0066] Figure 33 graphically depicts the effects of RIAA/ 'Acacia (1:5) supplementation (3 tablets per day) on fasting and 2 h post-prandial (pp) insulin levels. For the 2 h pp insulin level assessment, subjects presented after a 10-12 h fast and consumed a solution containing 75 g glucose (Trutol 100, CASCO NERL® Diagnostics); 2 h after the glucose challenge, blood was drawn and assayed for insulin levels (Laboratories Northwest, Tacoma, WA).
[0067] Figure 34 graphically depicts the effects of RΪAA/Acacia (1 :5) supplementation (3 tablets per day) on fasting and 2 h pp glucose levels. For the 2 h pp glucose level assessment, subjects presented after a 10-12 h fast and consumed a solution containing 75 g glucose (Trutol 100, CASCO NERL® Diagnostics); 2 h after the glucose challenge, blood was drawn and assayed for glucose levels (Laboratories Northwest, Tacoma, WA).
[0068] Figure 35 graphically depicts the effects of BIAA/Acacia (1 :5) supplementation (3 tablets per day) on HOMA scores. HOMA score was calculated from fasting insulin and glucose by published methods [(insulin (mcIU/mL)*glucose (mg/dL))/405]. [0069] Figure 36 graphically depicts the effects of BJAAJAcacia (1:5) supplementation (3 tablets per day) on serum TG levels.
[0070J Figure 37. Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
Cancer Cells by RIAA or Celecoxib-.Curcumin (1 :3).
[0071] Figure 38. Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
Cancer Cells by IAA, Celecoxib-.Curcumin (1 :3), or LY294002.
[0072] Figure 39. Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
Cancer Cells by THIAA or Celecoxib:Curcumin (1:3).
[0073] Figure 40. Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
Cancer Cells by HHIAA and Celecoxib-.Curcumin (1:3).
[00741 Figure 41. Percent Inhibition of (A) HT-29, (B) Caco-2 or (C) SW480 Colon
Cancer Cells by XN or Celecoxib-.Curcumin (1:3).
[0075] Figure 42. Observed and Expected Inhibition of (A) HT-29, (B) Caco-2 or (C)
S W480 Colon Cancer Cells by Combinations of Celecoxib and RIAA.
[0076] Figure 43. Observed and Expected Inhibition of (A) HT-29, (B) Caco-2 or (C)
SW480 Colon Cancer Cells by Combinations of Celecoxib and THIAA.
[0077] Figure 44 graphically displays the detection of THIAA in the serum over time following ingestion of 940 mg of THIAA.
[0078] Figure 45 displays the profile of THIAA detectable in the serum versus contol.
[0079] Figure 46 depicts the metabolism of THIAA by C YP2C9* 1.
DETAILED DESCRIPTION OF THE INVENTION
[0080] The present invention relates generally to methods and compositions that can be used to treat or inhibit cancers susceptible to protein kinase modulation. More specifically, the invention relates to methods and compositions which utilize compounds or derivatives commonly isolated either from hops or from members of the plant genus Acacia, or combinations thereof. 10081] The patents, published applications, and scientific literature referred to herein establish the knowledge of those with skill in the art and are hereby incorporated by reference in their entirety to the same extent as if each was specifically and individually indicated to be incorporated by reference. Any conflict between any reference cited herein and the specific teachings of this specification shall be resolved in favor of the latter. Likewise, any conflict between an art-understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this specification shall be resolved in favor of the latter.
[0082] Technical and scientific terms used herein have the meaning commonly understood by one of skill in the art to which the present invention pertains, unless otherwise defined. Reference is made herein to various methodologies and materials known to those of skill in the art. Standard reference works setting forth the general principles of recombinant DNA technology include Sambrook et ah, Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, New York (1989); Kaufman et al., Eds., Handbook of Molecular and Cellular Methods in Biology in Medicine, CRC Press, Boca Raton (1995); McPherson, Ed., Directed Mutagenesis: A Practical Approach, IRL Press, Oxford (1991). Standard reference works setting forth the general principles of pharmacology include Goodman and Gilman's The Pharmacological Basis of Therapeutics, 1 lth Ed., McGraw Hill Companies Inc., New York (2006).
[0083] In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. As used in this specification, the singular forms "a," "an" and "the" specifically also encompass the plural forms of the terms to which they refer, unless the content clearly dictates otherwise. Additionally, as used herein, unless specifically indicated otherwise, the word "or" is used in the "inclusive" sense of "and/or" and not the "exclusive" sense of "either/or." The term "about" is used herein to mean approximately, in the region of, roughly, or around. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term "about" is used herein to modify a numerical value above and below the stated value by a variance of 20%.
[0084] As used herein, the recitation of a numerical range for a variable is intended to convey that the invention may be practiced with the variable equal to any of the values within that range. Thus, for a variable which is inherently discrete, the variable can be equal to any integer value of the numerical range, including the end-ooints of the range. Similarly, for a variable which is inherently continuous, the variable can be equal to any real value of the numerical range, including the end-points of the range. As an example, a variable which is described as having values between 0 and 2, can be 0, 1 or 2 for variables which are inherently discrete, and can be 0.0, 0.1, 0.01, 0.001, or any other real value for variables which are inherently continuous.
(0085] Reference is made hereinafter in detail to specific embodiments of the invention. While the invention will be described in conjunction with these specific embodiments, it will be understood that it is not intended to limit the invention to such specific embodiments. On the contrary, it is intended to cover alternatives, modifications, and equivalents as may be included within the spirit and scope of the invention as defined by the appended claims. In the following description, numerous specific details are set forth in order to provide a thorough understanding of the present invention. The present invention may be practiced without some or all of these specific details. In other instances, well known process operations have not been described in detail, in order not to unnecessarily obscure the present invention.
[0086] Any suitable materials and/or methods known to those of skill can be utilized in carrying out the present invention. However, preferred materials and methods are described. Materials, reagents and the like to which reference are made in the following description and examples are obtainable from commercial sources, unless otherwise noted.
[0087] A first embodiment of the invention discloses methods to treat a cancer responsive to protein kinase modulation in a mammal in need where the method comprises administering to the mammal a therapeutically effective amount of a hexahydro-isoalpha acid. In some aspects of this embodiment, the hexahydro-isoalpha acid is selected from the group consisting of hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro- adhumulone.
[0088] In yet other aspects of this embodiment, the protein kinase modulated is selected from the group consisting of Abl(T315I), Aurora- A, Bmx, CDK9/cyclin Tl, CKI7I, CKlγ2, CKI73, cSRC, DAPKl, DAPK2, EphBl, ErbB4, Fer, FGFR2, GSK3β, GSK3α, HBPK3, IGF-IR, MAPKAP-K2, MSK2, PAK3, PAK5, PI3K, Pim-1, PKA(b), PKBB, PKB7, PRAK, Rsk2, Syk, Tie2, TrkA, TrkB, and ZEPK. [0089] In still other aspects the cancer responsive to kinase modulation is selected from the group consisting of bladder, breast, cervical, colon, lung, lymphoma, melanoma, prostate, thyroid, and uterine cancer.
[0090] Compositions used in the methods of this embodiment may further comprise one or more members selected from the group consisting of antioxidants, vitamins, minerals, proteins, fats, and carbohydrates, or a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents.
[0091] As used herein, "disease associated kinase" means those individual protein kinases or groups or families of kinases that are either directly causative of the disease or whose activation is associated with pathways which serve to exacerbate the symptoms of the disease in question.
[0092] The phrase "protein kinase modulation is beneficial to the health of the subject" refers to those instances wherein the kinase modulation (either up or down regulation) results in reducing, preventing, and/or reversing the symptoms of the disease or augments the activity of a secondary treatment modality.
[0093] The phrase "a cancer responsive to protein kinase modulation" refers to those instances where administration of the compounds of the invention either a) directly modulates a kinase in the cancer cell where that modulation results in an effect beneficial to the health of the subject (e.g., apoptosis or growth inhibition of the target cancer cell; b) modulates a secondary kinase wherein that modulation cascades or feeds into the modulation of a kinase which produces an effect beneficial to the health of the subject; or c) the target kinases modulated render the cancer cell more susceptible to secondary treatment modalities (e.g., chemotherapy or radiation therapy).
[0094] As used in this specification, whether in a transitional phrase or in the body of the claim, the terms "comprise(s)" and "comprising" are to be interpreted as having an open- ended meaning. That is, the terms are to be interpreted synonymously with the phrases "having at least" or "including at least". When used in the context of a process, the term "comprising" means that the process includes at least the recited steps, but may include additional steps. When used in the context of a compound or composition, the term "comprising" means that the compound or composition includes at least the recited features or compounds, but may also include additional features or compounds.
[0095] 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.
[0096] 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. "Spent hops" refers to the hops plant products remaining following a hops extraction procedure. See Verzele, M. and De Keukeleire, D., Developments in Food Science 27: Chemistry and Analysis of Hop and Beer Bitter Acids, Elsevier Science Pub. Co., 1991, New York, USA, herein incorporated by reference in its entirety, for a detailed discussion of hops chemistry. As used herein when in reference to a RIAA, "Rho" refers to those reduced isoalpha acids wherein the reduction is a reduction of the carbonyl group in the 4-methyl-3-pentenoyl side chain.
[0097] 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.
[0098] 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 subsequent removal of the CO2.
[0099] The term "pharmaceutically acceptable" is used in the sense of being compatible with the other ingredients of the compositions and not deleterious to the recipient thereof.
[00100] As used herein, "compounds" may be identified either by their chemical structure, chemical name, or common name. When the chemical structure and chemical or common name conflict, the chemical structure is determinative of the identity of the compound. The compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers. Accordingly, the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated or identified compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures. Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the skilled artisan. The compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated or identified compounds. The compounds described also encompass isotopically labeled compounds where one or more atoms have an atomic mass different from the atomic mass conventionally found in nature. Examples of isotopes that may be incorporated into the compounds of the invention include, but are not limited to, 2H, 3H, 13C, 14C, 15N, 18O5 17O, etc. Compounds may exist in unsolvated forms as well as solvated forms, including hydrated forms and as N-oxides. In general, compounds may be hydrated, solvated or N-oxides. Certain compounds may exist in multiple crystalline or amorphous forms. Also contemplated within the scope of the invention are congeners, analogs, hydrolysis products, metabolites and precursor or prodrugs of the compound. In general, unless otherwise indicated, all physical forms are equivalent for the uses contemplated herein and are intended to be within the scope of the present invention.
[00101] Compounds according to the invention may be present as salts. In particular, pharmaceutically acceptable salts of the compounds are contemplated. A "pharmaceutically acceptable salt" of the invention is a combination of a compound of the invention and either an acid or a base that forms a salt (such as, for example, the magnesium salt, denoted herein as "Mg" or "Mag") with the compound and is tolerated by a subject under therapeutic conditions. In general, a pharmaceutically acceptable salt of a compound of the invention will have a therapeutic index (the ratio of the lowest toxic dose to the lowest therapeutically effective dose) of 1 or greater. The person skilled in the art will recognize that the lowest therapeutically effective dose will vary from subject to subject and from indication to indication, and will thus adjust accordingly. [00102] As used herein "hop" or "hops" refers to plant cones of the genus Humulus which contain a bitter aromatic oil which is used in the brewing industry to prevent bacterial action and add the characteristic bitter taste to beer. More preferably, the hops used are derived from Humulus lupulus.
[00103] The term "acacia", as used herein, refers to any member of leguminous trees and shrubs of the genus Acacia. Preferably, the botanical compound derived from acacia is derived from Acacia catechu or Acacia nilotica.
[00104] The compounds according to the invention are optionally formulated in a pharmaceutically acceptable vehicle with any of the well known pharmaceutically acceptable carriers, including diluents and excipients (see Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, Mack Publishing Co., Easton, PA 1990 and Remington: The Science and Practice of Pharmacy, Lippincott, Williams & Wilkins, 1995). While the type of pharmaceutically acceptable carrier/vehicle employed in generating the compositions of the invention will vary depending upon the mode of administration of the composition to a mammal, generally pharmaceutically acceptable carriers are physiologically inert and nontoxic. Formulations of compositions according to the invention may contain more than one type of compound of the invention), as well any other pharmacologically active ingredient useful for the treatment of the symptom/condition being treated.
[00105] The term "modulate" or "modulation" is used herein to mean the up or down regulation of expression or activity of the enzyme by a compound, ingredient, etc., to which it refers.
[00106] As used herein, the term "protein kinase" represent transferase class enzymes that are able to transfer a phosphate group from a donor molecule to an amino acid residue of a protein. See Kostich, M., et ah, Human Members of the Eukaryotic Protein Kinase Family, Genome Biology 3(9):research0043.1-0043.12, 2002 herein incorporated by reference in its entirety, for a detailed discussion of protein kinases and family/group nomenclature.
[00107] Representative, non-limiting examples of kinases include AbI, Abl(T315I),
ALK, ALK4, AMPK, Arg, Arg, ARK5, ASKl, Aurora-A, AxI, BIk, Bmx, BRK, BrSKl, BrSK2, BTK, CaMKI, CaMKII, CaMKIV, CDKl/cyclinB, CDK2/cyclinA, CDK2/cyclinE, CDK3/cyclinE, CDK5/p25, CDK5/p35, CDK6/cyclinD3, CDK7/cyclinH/MATl, CDK9/cyclin Tl, CHKl, CHK2, CKl (y), CKl δ, CK2, CK2α2, cKit(D8l6V), cKit, c-RAF, CSK, cSRC, DAPKl, DAPK2, DDR2, DMPK, DRAKl5 DYRK2, EGFR, EGFR(L858R), EGFR(L861Q), EphAl, EphA2, EρhA3, EphA4, EphA5, EphA7, EphAS, EphBl, EphB2, EphB3, EphB4, ErbB4, Fer, Fes, FGFRl, FGFR2, FGFR3, FGFR4, Fgr, FItI, Flt3(D835Y), Flt3, FU4, Fms, Fyn, GSK3B, GSK3α, Hck, HIPKl, HIPK2, HEPK3, IGF-IR, HCKβ, IKKo, IR, IRAKI, IRAK4, IRR, ITK , JAK2, JAK3, JNKlαl, JTNK2o2, JNK3, KDR, Lck, LIMKl, LKBl, LOK3 Lyn, Lyn, MAPKl, MAPK2, MAPK2, MAPKAP-K2, MAPKAP-K3, MARKl, MEKl, MELK, Met, MINK, MKK4, MKK6, MKK7B, MLCK, MLKl, Mnk2, MRCKB, MRCKo, MSKl, MSK2, MSSKl, MSTl, MST2, MST3, MuSK, NEK2, NEK3, NEK6, NEK7, NLK , ρ70S6K, PAK2, PAK3, PAK4, PAK6, PAR-IBo; PDGFRβ, PDGFRo, PDKl, PI3K beta, PI3K delta, PI3K gamma, Pim-1, Pim-2. PKA(b), PKA, PKBB, PKBας PKBγ, PKCμ, PKCBI, PKCBH, PKCo, PKC7, PKCδ, PKCe, PKCf, PKCT?, PKC0, PKCi, PKD2, PKGlB, PKGlα, Plk3, PRAK, PRK2, PrKX, PTK5, Pyk2, Ret, RIPK2, ROCK-I, ROCK-II, ROCK-II, Ron, Ros, Rse, Rskl, Rskl, Rsk2, Rsk3, SAPK2a, SAPK2a(T106M), SAPK2b, SAPK3, SAPK4, SGK, SGK2, SGK3, SIK, Snk, SRPKl, SRPK2, STK33, Syk, TAKl, TBKl, Tie2, TrkA, TrkB, TSSKl, TSSK2, WNK2, WNK3, Yes, ZAP-70, ZIPK. In some embodiments, the kinases may be ALK, Aurora- A, AxI, CDK9/cyclin Tl, DAPKl, DAPK2, Fer, FGFR4, GSK3B, GSK3α, Hck, JNK2o2, MSK2, p70S6K, PAK3, PI3K delta, PI3K gamma, PKA, PKBB, PKBo, Rse, Rsk2, Syk, TrkA, and TSSKl. In yet other embodiments the kinase is selected from the group consisting of ABL, AKT, AURORA, CDK, DBF2/20, EGFR, EPH/ELK/ECK, ERK/MAPKFGFR, GSK3, IKKB, BSfSR, JAK DOM 1/2, MARK/PRKAA, MEK/STE7, MEKK/STE11, MLK, mTOR, PAK/STE20, PDGFR, PI3K, PKC, POLO, SRC, TEC/ATK, and ZAP/SYK.
[00108] The methods and compositions of the present invention are intended for use with any mammal that may experience the benefits of the methods of the invention. Foremost among such mammals are humans, although the invention is not intended to be so limited, and is applicable to veterinary uses. Thus, in accordance with the invention, "mammals" or "mammal in need" include humans as well as non-human mammals, particularly domesticated animals including, without limitation, cats, dogs, and horses.
[00109] As used herein, "autoimmune disorder" refers to those diseases, illnesses, or conditions engendered when the host's systems are attacked by the host's own immune system. Representative, non-limiting examples of autoimmune diseases include alopecia areata, ankylosing spondylitis, arthritis, antiphospholipid syndrome, autoimmune Addison's disease, autoimmune hemolytic anemia, autoimmune inner ear disease (also known as Meniers disease), autoimmune lymphoproliferative syndrome (ALPS), autoimmune thrombocytopenic purpura, autoimmune hemolytic anemia, autoimmune hepatitis, Bechet's disease, Crohn's disease, diabetes mellitus type 1, glomerulonephritis, Graves' disease, Guillain-Barre syndrome, inflammatory bowel disease, lupus nephritis, multiple sclerosis, myasthenia gravis, pemphigus, pernicious anemia, polyarteritis nodosa, polymyositis, primary billiary cirrhosis, psoriasis, rheumatic fever, rheumatoid arthritis, scleroderma, Sjogren's syndrome, systemic lupus erythematosus, ulcerative colitis, vitiligo, and Wegener's granulamatosis. Representative, non-limiting examples of kinases associated with autoimmune disorders include AMPK, BTK, ERK, FGFR5 FMS, GSK, IGFR, IKK, JAK, PDGFR, PDK, PKC, PLK, ROCK, and VEGFR.
[00110] "Allergic disorders", as used herein, refers to an exaggerated or pathological reaction (as by sneezing, respiratory distress, itching, or skin rashes) to substances, situations, or physical states that are without comparable effect on the average individual. As used herein, "inflammatory disorders" means a response (usually local) to cellular injury that is marked by capillary dilatation, leukocytic infiltration, redness, heat, pain, swelling, and often loss of function and that serves as a mechanism initiating the elimination of noxious agents and of damaged tissue. Examples of allergic or inflammatory disorders include, without limitation, asthma, rhinitis, ulcerative colitis, Crohn's disease, pancreatitis, gastritis, benign tumors, polyps, hereditary polyposis syndrome, colon cancer, rectal cancer, breast cancer, prostate cancer, stomach cancer, ulcerous disease of the digestive organs, stenocardia, atherosclerosis, myocardial infarction, sequelae of stenocardia or myocardial infarction, senile dementia, and cerebrovascular diseases. Representative, non-limiting examples of kinases associated with allergic disorders include AKT, AMPK, BTK, CHK, EGFR, FYN, IGF-IR, IKKB, ITK, JAK, KIT, LCK, LYN, MAPK, MEK, mTOR, PDGFR, PBK, PKC, PPAR, ROCK, SRC, SYK, and ZAP.
[00111] As used herein, "metabolic syndrome" and "diabetes associated disorders" refers to insulin related disorders, i.e., to those diseases or conditions where the response to insulin is either causative of the disease or has been implicated in the progression or suppression of the disease or condition. Representative examples of insulin related disorders include, without limitation diabetes, diabetic complications, insulin sensitivity, polycystic ovary disease, hyperglycemia, dyslipidemia, insulin resistance, metabolic syndrome, obesity, body weight gain, inflammatory diseases, diseases of the digestive organs, stenocardia, myocardial infarction, sequelae of stenocardia or myocardial infarction, senile dementia, and cerebrovascular dementia. See, Harrison's Principles of Internal Medicine, 16h Ed., McGraw Hill Companies Inc., New York (2005). Examples, without limitation, of inflammatory conditions include diseases of the digestive organs (such as ulcerative colitis, Crohn's disease, pancreatitis, gastritis, benign tumor of the digestive organs, digestive polyps, hereditary polyposis syndrome, colon cancer, rectal cancer, stomach cancer and ulcerous diseases of the digestive organs), stenocardia, myocardial infarction, sequelae of stenocardia or myocardial infarction, senile dementia, cerebrovascular dementia, immunological diseases and cancer in general. Non-limiting examples of kinases associated with metabolic syndrome can include AKT, AMPK, CDK, CSK, ERK, GSK, IGFR, JNK, MAPK, MEK, PBK, and PKC.
[00112] "Insulin resistance" refers to a reduced sensitivity to insulin by the body's insulin-dependent processes resulting in lowered activity of these processes or an increase in insulin production or both. Insulin resistance is typical of type 2 diabetes but may also occur in the absence of diabetes.
[00113] As used herein "diabetic complications" include, without limitation, retinopathy, muscle infarction, idiopathic skeletal hyperostosis and bone loss, foot ulcers, neuropathy, arteriosclerosis, respiratory autonomic neuropathy and structural derangement of the thorax and lung parenchyma, left ventricular hypertrophy, cardiovascular morbidity, progressive loss of kidney function, and anemia.
[00114] As used herein "cancer" refers to any of various benign or malignant neoplasms characterized by the proliferation of anaplastic cells that, if malignant, tend to invade surrounding tissue and metastasize to new body sites. Representative, non-limiting examples of cancers considered within the scope of this invention include brain, breast, colon, kidney, leukemia, liver, lung, and prostate cancers. Non-limiting examples of cancer associated protein kinases considered within the scope of this invention include ABL, AKT, AMPK, Aurora, BRK, CDK, CHK, EGFR, ERB, FGFR, IGFR, KIT, MAPK, mTOR, PDGFR, PBK, PKC, and SRC.
[00115] "Ocular disorders", refers to those disturbances in the structure or function of the eye resulting from developmental abnormality, disease, injury, age or toxin. Non-limiting examples of ocular disorders considered within the scope of the present invention include retinopathy, macular degeneration or diabetic retinopathy. Ocular disorder associated kinases include, without limitation, AMPK, Aurora, EPH, ERB, ERK, FMS, IGFR, MEK, PDGFR, PI3K, PKC, SRC, and VEGFR.
[00116] A "neurological disorder", as used herein, refers to any disturbance in the structure or function of the central nervous system resulting from developmental abnormality, disease, injury or toxin. Representative, non-limiting examples of neurological disorders include Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS or Lou Gehrig's Disease), Huntington's disease, neurocognitive dysfunction, senile dementia, and mood disorder diseases. Protein kinases associated with neurological disorders may include, without limitation, AMPK, CDK, FYN, JNK, MAPK, PKC, ROCK, RTK, SRC, and VEGFR.
[00117] As used herein "cardiovascular disease" or "CVD" refers to those pathologies or conditions which impair the function of, or destroy cardiac tissue or blood vessels. Cardiovascular disease associated kinases include, without limitation, AKT, AMPK, GRK5 GSK, IGF-IR, DCKB, JAK, JUN, MAPK, PKC, RHO, ROCK, and TOR.
[00118] "Osteoporosis", as used herein, refers to a disease in which the bones have become extremely porous, thereby making the bone more susceptible to fracture and slower healing. Protein kinases associated with osteoporosis include, without limitation, AKT, AMPK, CAMK, IRAK-M, MAPK, mTOR, PPAR, RHO, ROS, SRC, SYR, and VEGFR.
[00119] An embodiment of the invention describes compositions to treat a cancer responsive to protein kinase modulation in a mammal in need. The compositions comprise a therapeutically effective amount of a hexahydro-isoalpha acid; wherein the therapeutically effective amount modulates a cancer associated protein kinase. In some aspects of this embodiment, the hexahydro-isoalpha acid is selected from the group consisting of hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.
[00120] In other aspects of this embodiment, the compositions further comprise a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents. [00121] In yet other aspects, the compositions further comprise one or more members selected from the group consisting of antioxidants, vitamins, minerals, proteins, fats, and carbohydrates.
[00122] As used herein, by "treating" is meant reducing, preventing, and/or reversing the symptoms in the individual to which a compound of the invention has been administered, as compared to the symptoms of an individual not being treated according to the invention. A practitioner will appreciate that the compounds, compositions, and methods described herein are to be used in concomitance with continuous clinical evaluations by a skilled practitioner (physician or veterinarian) to determine subsequent therapy. Hence, following treatment the practitioners will evaluate any improvement in the treatment of the pulmonary inflammation according to standard methodologies. Such evaluation will aid and inform in evaluating whether to increase, reduce or continue a particular treatment dose, mode of administration, etc.
[00123] It will be understood that the subject to which a compound of the invention is administered need not suffer from a specific traumatic state. Indeed, the compounds of the invention may be administered prophylactically, prior to any development of symptoms. The term "therapeutic," "therapeutically," and permutations of these terms are used to encompass therapeutic, palliative as well as prophylactic uses. Hence, as used herein, by "treating or alleviating the symptoms" is meant reducing, preventing, and/or reversing the symptoms of the individual to which a compound of the invention has been administered, as compared to the symptoms of an individual receiving no such administration.
[00124] The term "therapeutically effective amount" is used to denote treatments at dosages effective to achieve the therapeutic result sought. Furthermore, one of skill will appreciate that the therapeutically effective amount of the compound of the invention may be lowered or increased by fine tuning and/or by administering more than one compound of the invention, or by administering a compound of the invention with another compound. See, for example, Meiner, C.L., "Clinical Trials: Design, Conduct, and Analysis," Monographs in Epidemiology and Biostatistics, Vol. 8 Oxford University Press, USA (1986). The invention therefore provides a method to tailor the administration/treatment to the particular exigencies specific to a given mammal. As illustrated in the following examples, therapeutically effective amounts may be easily determined for example empirically by starting at relatively low amounts and by step-wise increments with concurrent evaluation of beneficial effect. [00125] It will be appreciated by those of skill in the art that the number of administrations of the compounds according to the invention will vary from patient to patient based on the particular medical status of that patient at any given time including other clinical factors such as age, weight and condition of the mammal and the route of administration chosen.
[00126] As used herein, "symptom" denotes any sensation or change in bodily function that is experienced by a patient and is associated with a particular disease, i.e., anything that accompanies "X" and is regarded as an indication of "X'"s existence. It is recognized and understood that symptoms will vary from disease to disease or condition to condition. By way of non-limiting examples, symptoms associated with autoimmune disorders include fatigue, dizziness, malaise, increase in size of an organ or tissue (for example, thyroid enlargement in Grave's Disease), or destruction of an organ or tissue resulting in decreased functioning of an organ or tissue (for example, the islet cells of the pancreas are destroyed in diabetes).
[00127] Representative symptomology for allergy associated diseases or conditions include absentmindedness, anaphylaxis, asthma, burning eyes, constipation, coughing, dark circles under or around the eyes, dermatitis, depression, diarrhea, difficulty swallowing, distraction or difficulty with concentration, dizziness, eczema, embarrassment, fatigue, flushing, headaches, heart palpitations, hives, impaired sense of smell, irritability/behavioral problems, itchy nose or skin or throat, joint aches muscle pains, nasal congestion, nasal polyps, nausea, postnasal drainage (postnasal drip), rapid pulse, rhinorrhea (runny nose), ringing - popping or fullness in the ears, shortness of breath, skin rashes, sleep difficulties, sneezing, swelling (angioedema), throat hoarseness, tingling nose, tiredness, vertigo, vomiting, watery or itchy or crusty or red eyes, and wheezing.
[00128] "Inflammation" or "inflammatory condition" as used herein refers to a local response to cellular injury that is marked by capillary dilatation, leukocytic infiltration, redness, heat, pain, swelling, and often loss of function and that serves as a mechanism initiating the elimination of noxious agents and of damaged tissue. Representative symptoms of inflammation or an inflammatory condition include, if confined to a joint, redness, swollen joint that's warm to touch, joint pain and stiffness, and loss of joint function. Systemic inflammatory responses can produce "flu-like" symptoms, such as, for instance, fever, chills, fatigue/loss of energy, headaches, loss of appetite, and muscle stiffness. [00129] Diabetes and metabolic syndrome often go undiagnosed because many of their symptoms seem so harmless. For example, some diabetes symptoms include, without limitation: frequent urination, excessive thirst, extreme hunger, unusual weight loss, increased fatigue, irritability, and blurry vision.
[00130] Symptomology of neurological disorders may be variable and can include, without limitation, numbness, tingling, hyperesthesia (increased sensitivity), paralysis, localized weakness, dysarthria (difficult speech), aphasia (inability to speak), dysphagia (difficulty swallowing), diplopia (double vision), cognition issues (inability to concentrate, for example), memory loss, amaurosis fugax (temporary loss of vision in one eye) difficulty walking, incoordination, tremor, seizures, confusion, lethargy, dementia, delirium and coma.
[00131] The following examples are intended to further illustrate certain preferred embodiments of the invention and are not limiting in nature. Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific substances and procedures described herein.
EXAMPLES
Example 1
Effects of modified Hops components on protein kinases
[00132] As stated above, kinases represent transferase class enzymes that are able to transfer a phosphate group from a donor molecule (usually ATP) to an amino acid residue of a protein (usually threonine, serine or tyrosine). Kinases are used in signal transduction for the regulation of enzymes, i.e., they can inhibit or activate the activity of an enzyme, such as in cholesterol biosynthesis, amino acid transformations, or glycogen turnover. While most kinases are specialized to a single kind of amino acid residue, some kinases exhibit dual activity in that they can phosphorylate two different kinds of amino acids. As shown in Figure 1, kinases function in signal transduction and translation.
[00133] Methods - The inhibitory effect of 10 μg RIAA/ml of the present invention on human kinase activity was tested on a panel of over 200 kinases in the KinaseProfiler™ Assay (Upstate Cell Signaling Solutions, Upstate USA, Inc., Charlottes vi lie, VA., USA). The assay protocols for specific kinases are summarized at http://www.upstate.com/img/pdfykp protocols_full.pdf (last visited on June 12, 2006V
[00134] Results - Just over 205 human kinases were assayed in the cell free system.
Surprisingly we discovered that the hops compounds tested inhibited 25 of the 205 kinases by 10% or greater. Eight (8) of the 205 were inhibited by >20%; 5 of 205 were inhibited by >30; and 2 were inhibited by about 50%.
[00135] Specifically in the PBkinase pathway, hops inhibits PI3Kγ, PI3Kδ, PI3Kβ,
Aktl, Akt2, GSK3α, GSK3β, P70S6K. It should be noted that mTOR was not available for testing.
[00136] The inhibitory effects of the hops compounds RIAA on the kinases tested are shown in Table 1 below.
Table 1
Kinase inhibition by RIAA tested in the KinasePro filer™ Assay at 10 uu/ml
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
[00137] It should be noted that several kinases in the PI3K pathway are being preferentially inhibited by RIAA, for example, Aktl at 51% inhibition. It is interesting to note that three Akt isoforms exist. Aktl null mice are viable, but retarded in growth [Cho et al., Science 292:1728-1731 (2001)]. Drosophila eye cells deficient in Aktl are reduced in size [Verdu et al., Nat cell Biol 1 :500-505 (1999)]; overexpression leads to increased size from normal. Akt2 null mice are viable but have impaired glucose control [Cho et al., J Biol Chem 276:38345-38352 (2001)]. Hence, it appears Aktl plays a role in size determination and Akt2 is involved in insulin signaling. [00138] The PDK pathway is known to play a key role in mRNA stability and mRNA translation selection resulting in differential protein expression of various oncogene proteins and inflammatory pathway proteins. A particular 5' mRNA structure denoted 5'-TOP has been shown to be a key structure in the regulation of mRNA translation selection.
[00139] A review of the cPLA literature and DNA sequence indicates that the 5' mRNA of human cPLA2 contains a consensus (82% homology to a known oncogene regulated similarly) sequence indicating that it too has a 5'TOP structure. sPLAs, also known to be implicated in inflammation, also have this same 5'-TOP. Moreover, this indicates that cPLA2 and possibly other PLAs are upregulated by the PI3K pathway via increasing the translation selection of cPLA2 mRNA resulting in increases in cPLA2 protein. Conversely, inhibitors of PI3K should reduce the amount of cPLA2 and reduce PGE2 formation made via the C0X2 pathway.
[00140] Taken together the kinase data and our own results where we have discovered that hops compounds inhibit cPLA2 protein expression (Western blots, data not shown) but not mRNA, suggests that the anti-inflammatory mode of action of hops compounds may be via reducing substrate availability to COX2 by reducing cPLA2 protein levels, and perhaps more specifically, by inhibiting the PI3K pathway resulting in the inhibition of activation of TOP mRNA translation.
[00141] The exact pathway of activity remains unclear. Some reports are consistent with the model that activation occurs via phosphorylation of one or more of the six isoforms of ribosomal protein S6 (RPS6). RPS6 is reported to resolve the 5'TOP mRNA allowing efficient translation into protein. However, Stolovich et al. MoI Cell Biol Dec, 8101-81 13 (2002), disputes this model and proposes that Aktl phosphorylates an unknown translation factor, X, which allows TOP mRNA translation.
Example 2
Dose response effects of hops or acacia components on selected protein kinases
[00142] The dose responsiveness of mgRho was tested at approximately 10, 50, and
100 μg/ml on over sixty selected protein kinases according to the protocols of Example 1 are presented as Tables 2A & 2B below. The five kinases which were inhibited the most are displayed graphically as Figure 2. [00143] The dose responsiveness for kinase inhibition (reported as a percent of control) of a THIAA preparation was tested at approximately 1, 10, 25, and 50 ug/ml on 86 selected kinases as presented in Table 3 below. Similarly, an acacia preparation was tested at approximately 1, 5, and 25 ug/ml on over 230 selected protein kinases according to the protocols of Example 1 and are presented as Table 4 below. Preparations of isoalpha acids (IAA), heaxahydroisoalpha acids (HHIAA), beta acids, and xanthohumol were also tested at approximately 1, 10, 25, and 50 ug/ml on 86 selected kinases and the dose responsiveness results are presented below as Tables 5 — 8 respectively.
Table 2A
Dose response effect (as % of Control) of a mgRho on selected protein kinases
Figure imgf000034_0001
Figure imgf000035_0001
Table 2B
Dose response effect (as % of Control) of a mgRho on selected protein kinases
Figure imgf000035_0002
Figure imgf000036_0001
Table 3
Dose response effect (as % of Control) of THIAA on selected protein kinases
Figure imgf000036_0002
Figure imgf000037_0001
Figure imgf000038_0001
Table 4
Dose response effect fas % of Control) of acacia on selected protein kinases
Figure imgf000038_0002
Figure imgf000039_0001
Figure imgf000040_0001
Table 5
Dose response effect fas % of Control) of IAA on selected protein kinases
Figure imgf000040_0002
Figure imgf000041_0001
Figure imgf000042_0001
Table 6
Dose response effect (as % of Control") of HHIAA on selected protein kinases
Figure imgf000042_0002
Figure imgf000043_0001
Figure imgf000044_0001
Table 7
Dose response effect (as % of Control) of beta acids on selected protein kinases
Figure imgf000044_0002
Figure imgf000045_0001
Figure imgf000046_0001
Table 8
Dose response effect (as % of ControD of xanthohumol on selected protein kinases
Figure imgf000046_0002
Figure imgf000047_0001
Figure imgf000048_0001
[00144J Results — The effect on kinase activity modulation by the various compounds tested displayed a wide range of modulatory effects depending on the specific kinase and compound tested (Tables 2 - 8) with representative examples enumerated below.
[00145] PI3Kδ, a kinase strongly implicated in autoimmune diseases such as, for example, rheumatoid arthritis and lupus erythematosus, exhibited a response inhibiting 36%, 78% and 87% of kinase activity at 10, 50, and 100 ug/ml respectively for MgRho. MgRho inhibited Syk in a dose dependent manner with 21%, 54% and 72% inhibition at 10, 50, and 100 μg/ml respectively. Additionally, GSK or glycogen synthase kinase (both GSK alpha and beta) displayed inhibition following mgRho exposure (alpha, 35, 36, 87% inhibition; beta, 35, 83, 74 % inhibition respectively at 10, 50, 100 μg/mi). See Table 2. [00146] THIAA displayed a dose dependent inhibition of kinase activity for many of the kinases examined with inhibition of FGFR2 of 7%, 16%, 77%, and 91% at 1, 5, 25, and 50 μg/ml respectively. Similar results were observed for FGFR3 (0%, 6%, 61%, and 84%) and TrkA (24%, 45%, 93%, and 94%) at 1, 5, 25, and 50 μg/ml respectively. See Table 3.
[00147] The acacia extract tested (A. niloticά) appeared to be the most potent inhibitor of kinase activity examined (Table 4), demonstrating 80% or greater inhibition of activity for such kinases as Syk (98%), Lyn (96%), GSK3α (95%), Aurora-A (92%), Flt4 (88%), MSSKl (88%), GSK3B (87%), BTK (85%), PRAK (82%), and TrkA (80%), all at a 1 μg/ml exposure.
Example 3
Effect of hops components on PI3K activity
[00148] The inhibitory effect on human PI3K-& PDK-7, and PI3K-δ of the hops components xanthohumol and the magnesium salts of beta acids, isoalpha acids (Mg-IAA), tetrahydro-isoalpha acids (Mg-THIAA), and hexahydro-isoalpha acids (Mg-HHIAA) were examined according to the procedures and protocols of Example 1. Additionally examined was an Acacia nilotica heartwood extract. All compounds were tested at 50 μg/ml. The results are presented graphically as Figure 3.
[00149] It should be noted that all of the hops compounds tested showed >50% inhibition of PI3K activity with Mg-THIAA producing the greatest overall inhibition (>80% inhibition for all PI3K isoforms tested). Further note that both xanthohumol and Mg-beta acids were more inhibitory to PI3K-γthan to PI3K-0 or PI3K-6. Mg-IAA was approximately 3-fold more inhibitory to PI3K-/? than to PI3K-γ or PDK-δ. The Acacia nilotica heartwood extract appeared to stimulate PI3K-/3 or PI3K-δ activity. Comparable results were obtained for Syk and GSK kinases (data not shown).
Example 4
Inhibition of PGE? synthesis in stimulated and nonstimmulated murine macrophages by hops compounds and derivatives
[00150] The objective of this example was to assess the extent to which hops derivatives inhibited COX-2 synthesis of PGE2 preferentially over COX-I synthesis of PGE2 in the murine RAW 264.7 macrophage model. The RAW 264.7 cell line is a well-established model for assessing anti-inflammatory activity of test agents. Stimulation of RAW 264.7 cells with bacterial lipopolysaccharide induces the expression of COX-2 and production of PGE2. Inhibition of PGE2 synthesis is used as a metric for anti-inflammatory activity of the test agent.. Equipment, Chemicals and Reagents, PGE2 assay, and calculations are described below.
[00151] Equipment - Equipment used in this example included an OHAS Model
#E01140 analytical balance, a Forma Model #F1214 biosafety cabinet (Marietta, Ohio), various pipettes to deliver 0.1 to 100 μl (VWR, Rochester, NY), a cell hand tally counter (VWR Catalog #23609-102, Rochester, NY), a Forma Model #F3210 CO2 incubator (Marietta, Ohio), a hemocytometer (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).
[00152] Chemicals and Reagents - Bacterial lipopolysaccharide (LPS; B E. coli
055 :B5) was from Sigma (St. Louis, MO). Heat inactivated Fetal Bovine Serum (FBS-HI Cat. #35-011CV), and Dulbecco's Modification of Eagle's Medium (DMEM Cat #10- 013CV) was purchased from Mediatech (Herndon, VA). 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), (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 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.
[00153] Test materials — Hops derivatives as described in Table 12 were used. The
COX-I selective inhibitor aspirin and COX-2 selective inhibitor celecoxib were used as positive controls. Aspirin was obtained from Sigma (St. Louis, MO) and the commercial formulation of celecoxib was used (Celebrex™, Searle & Co., Chicago, IL). [00154] Cell culture and treatment with test material — 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.
[00155] For COX-2 associated PGE2 synthesis, 100 μl of medium was removed from each well of the cell plates prepared on day one and replaced with 100 μl of equilibrated 2X final concentration of the test compounds. Cells were then incubated for 90 minutes. Twenty μl of LPS were added to each well of cells to be stimulated to achieve a final concentration of 1 μg LPS/ml and the cells were incubated for 4 h. The cells were further incubated with 5 μM arachadonic acid for 15 minutes. Twenty-five μl of supernatant medium from each well was transferred to a clean microfuge tube for the determination of PGE2 released into the medium.
[00156] For COX-I associated PGE2 synthesis, 100 μl of medium were removed from each well of the cell plates prepared on day one and replaced with 100 μl 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 μM arachadonic acid for 15 minutes. Twenty-five μl of supernatant medium from each well was transferred to a clean microfuge tube for the determination of PGE2 released into the medium.
[00157] The appearance of the cells was observed and viability was assessed visually.
No apparent toxicity was observed at the highest concentrations tested for any of the compounds. Twenty-five μl 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 below.
[00158] PGE2 assay - A commercial, non-radioactive procedure for quantification of
PGE2 was employed (Caymen Chemical, Ann Arbor, MI) and the recommended procedure of the manufacturer was used without modification. Briefly, 25 μl of the 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. After the wells were emptied and rinsed with wash buffer, 200 μl of Ellman's reagent containing substrate for acetylcholinesterase were added. The reaction was maintained on a slow shaker at room temperature for 1 h and the absorbance at 415 run was determined in a Bio-Tek Instruments (Model #Elx800, Winooski, VT) ELISA plate reader. The PGE2 concentration was represented as picograms per ml. 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'l. The median inhibitory concentrations (IC50) for PGE2 synthesis from both COX-2 and COX-I were calculated as described below.
[00159] Calculations - The median inhibitory concentrations (IC50) for PGE2 synthesis were calculated using CalcuSyn (BIOSOFT, Ferguson, MO). A minimum of four concentrations of each test material or positive control was used for computation. This statistical package performs multiple drug dose-effect calculations using the Median Effect methods described by T.C Chou and P. Talalay [Chou. T.C. and P. Talalay. Quantitative analysis of dose-effect relationships; the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 22: 27-55, (1984)] and is incorporated herein by reference. 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.
[00160] Median inhibitory concentrations were ranked into four arbitrary categories:
(1) highest anti-inflammatory response for those agents with an IC50 values within 0.3 μg/ml of 0.1; (2) high anti-inflammatory response for those agents with an IC50 value within 0.7 μg/ml of 1.0; (3) intermediate anti-inflammatory response for those agents with IC50 values between 2 and 7 μg/ml; and (4) low anti-inflammatory response for those agents with IC50 values greater than 12 μg/ml, the highest concentration tested
[001611 Results - The aspirin and celecoxib positive controls demonstrated their respective cyclooxygenase selectivity in this model system (Table 9). While aspirin was approximately 1000-fold more selective for COX-I, celecoxib was 114 times more selective for COX-2. All hops materials were COX-2 selective with Rho isoalpha acids and isoalpha acids demonstrating the highest COX-2 selectivity, 363- and 138-fold respectively. Such high COX-2 selectivity combined with low median inhibitory concentrations, has not been previously reported for natural products from other sources. Of the remaining hops derivatives, only the aromahop oil exhibited a marginal COX-2 selectivity of 3-fold. For extrapolating in vitro data to clinical efficacy, it is generally assumed that a COX-2 selectivity of 5-fold or greater indicates the potential for clinically significant protection of gastric mucosa. Under this criterion, beta acids, CO2 hop extract, spent hops CCVethanol, tetrahydro isoalpha acids and hexahydro isoalpha acids displayed potentially clinically relevant COX-2 selectivity.
Table 9
CQX-2 and COX-I inhibition in RAW 264.7 cells by hop fractions and derivatives
Figure imgf000053_0001
Example 5
Lack of direct PGE? inhibition by reduced isomerized alpha acids or isomerized alpha acids in LPS-stimulated Raw 264.7 cells
[00162] The objective of this study was to assess the ability of the hops derivatives reduced isoalpha acids and isomerized alpha acids to function independently as direct inhibitors of COX-2 mediated PGE2 biosynthesis in the RAW 264.7 cell model of inflammation. The RAW 264.7 cell line as described in Example 4 was used in this example. Equipment, chemicals and reagents, PGE2 assay, and calculations were as described in Example 4.
[00163] Test materials - Hops derivatives reduced isoalpha acids and isomerized alpha acids, as described in Table 12, were used. Aspirin, a COX-I selective positive control, was obtained from Sigma (St. Louis, MO). [00164] Cell culture and treatment with test material - RAW 264.7 cells (TIB-71) were obtained from the American Type Culture Collection (Manassas, VA) and sub-cultured as described in Example 4. Following overnight incubation at 37°C with 5% CO2, the growth medium was aspirated and replaced with 200 μl DMEM without FBS or penicillin/streptomycin. RAW 264.7 cells were stimulated with LPS and incubated overnight to induce COX-2 expression. Eighteen hours post LPS-stimulation, test materials were added followed 60 minutes later by the addition of the calcium ionophore A23187. Test materials were dissolved in DMSO as a 250-fold stock solution. Four μl of this 250-fold stock test material preparation was added to 1 ml of DMEM and 200 μl of this solution was subsequently added to eight wells for each dose of test material. Supernatant media was sampled for PGE2 determination after 30 minutes. Median inhibitory concentrations were computed from a minimum of four concentrations over two independent experiments as described in Example 4.
[00165] 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 as described in Example 4.
[00166] Cell viability — Cell viability was assessed by microscopic inspection of cells prior to or immediately following sampling of the medium for PGE2 assay. No apparent cell mortality was noted at any of the concentrations tested.
[00167] Calculations — Four concentrations 0.10, 1.0, 10 and 100 μg/ml were used to derive dose-response curves and compute medium inhibitory concentrations (ICsos) with 95% confidence intervals using CalcuSyn (BIOSOFT, Ferguson, MO).
[00168] Results - LPS-stimulation of PGE2 production in RAW 264.7 cells ranged from 1.4-fold to 2.1 -fold relative to non-stimulated cells. The IC50 value of 8.7 μg/ml (95% CL = 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 μg/ml [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. ScL USA 96:7563- 7568, (1999)] and historical data of this laboratory of 3.2 μg/ml (95% CL = 0.55 - 19) in the A549 cell line. [00169] When added following COX-2 induction in RAW 264.7 cells by LPS, both
RIAA and IAA produced only modest, dose-related inhibition Of PGE2. 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 10) in the mg/ml range for RIAA (36 mg/ml) and IAA (>1000 mg/ml). The minimal changes observed in response over three-log units of doses suggests that the observed PGE2 inhibitory effect of the hops derivatives in this cell-based assay may be a secondary effect on the cells and not a direct inhibition of COX-2 enzyme activity.
[001701 Figure 4A and 4B depict the dose-response data respectively, for RIAA and
IAA as white bars and the dose-response data from this example as gray bars. The effect of sequence of addition is clearly seen and supports the inference that RIAA and IAA are not direct COX-2 enzyme inhibitors.
[00171] It appears that (1) hop materials were among the most active, antiinflammatory natural products tested as assessed by their ability to inhibit PGE2 biosynthesis in vitro; (2) RIAA and IAA do not appear to be direct COX-2 enzyme inhibitors based on their pattern of inhibition with respect to COX-2 induction; and (3) RIAA and IAA have a COX-2 selectively that appears to be based on inhibition of COX-2 expression, not COX-2 enzyme inhibition. This selectivity differs from celecoxib, whose selectivity is based on differential enzyme inhibition.
Table 10
Median inhibitory concentrations for RIAA, IAA in RAW 264.7 cells when test material is added post overnight LPS-stimulation.
Figure imgf000055_0001
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.
Example 6
Hops compounds and derivatives are not direct cvclooxygenase enzyme inhibitors in A549 pulmonary epithelial cells
(00172] Chemicals — Hops and hops derivatives used in this example were previously described in Example 4. All other chemicals were obtained from suppliers as described in Example 4.
[00173] Equipment, PGE 2 assay, and Calculations were as described in Example 4.
[00174] 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 μg streptomycin/ml, 5 mM sodium pyruvate, and 5 mM L-glutamine. On the day of the experiments, exponentially growing cells were harvested and washed with serum-free RPMI 1640.
[0017S] Log phase A549 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 OfPGE2 inhibition by the test compounds, the procedure of Warner, et al. [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, (1999)], also known as the WHMA-COX-2 protocol was followed with no modification. Briefly, 24 hours after plating of the A549 cells, interleukin-lβ (10 ng/ml) was added to induce the expression of COX-2. After 24 hr, the cells were washed with serum-free RPMI 1640. Subsequently, 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 μg/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 μM) was added to the wells to release arachadonic acid. Twenty-five μl of media were sampled from the wells 30 minutes later for PGE2 determination. [00176] Cell viability was assessed visually and no apparent 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 4. The median inhibitory concentration (ICso) for PGE2 synthesis was calculated as previously described in Example 4.
[00177] Results — At the doses tested, the experimental protocol failed to capture a median effective concentration for any of the hops extracts or derivatives. Since the protocol requires the stimulation of COX-2 expression prior to the addition of the test compounds, it is believed that the failure of the test materials to inhibit PGE2 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 was observed using the WHMA-COX-2 protocol, this procedure appears inappropriate in evaluating the anti-inflammatory properties of hops compounds or derivatives of hops compounds.
Example 7
Hops derivatives inhibit mite dust allergen activation of PGE? biosynthesis in A549 pulmonary epithelial cells
[00178] Chemicals — Hops and hops derivatives, (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), used in this example were previously described in Example 1. All other chemicals were obtained from suppliers as described in Example 4. Test materials at a final concentration of 10 μg/ml were added 60 minutes prior to the addition of the mite dust allergen.
[00179] Equipment, PGE2 assay, and Calculations were as described in Example 4.
[00180] Mite dust allergen isolation - Dermatophagoides farinae is the American house dust mite. D. farinae were raised 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 μm 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.
J00181] Cell culture and treatment - The human airway epithelial cell line, A549
(American Type Culture Collection, Bethesda, MD) was cultured and treated as previously described in Example 6. Mite allergen was added to the culture medium to achieve a final concentration of 1000 ng/ml. Eighteen hours later, the media were sampled for PGE2 determination.
[00182] Results - Table 11 depicts the extent of inhibition by hops derivatives OfPGE2 biosynthesis in A549 pulmonary cells stimulated by mite dust allergen. All hops derivatives tested were capable of significantly inhibiting the stimulatory effects of mite dust allergens.
Table 11
PGE? inhibition by hops derivatives in A549 pulmonary epithelial cells stimulated by mite dust allergen.
Figure imgf000058_0001
[00183] This example illustrates that hops derivatives are capable of inhibiting the
PGE2 stimulatory effects of mite dust allergens in A549 pulmonary cells.
Example 8
Lack of Direct COX-2 Inhibition by Reduced Isoalpha Acids
[00184] The objective of this example was to determine whether magnesium reduced isoalpha acids can act as a direct inhibitor of COX-2 enzymatic activity. (00185] Materials - Test compounds were prepared in dimethyl sulfoxide (DMSO) and stored at -2O0C. LPS was purchased from Sigma-Aldrich (St. Louis, MO). MgRIAA was supplied by Metagenics (San Clemente, CA), and the commercial formulation of celecoxib was used (Celebrex™, Searle & Co., Chicago, IL).
[00186] Cell Culture — The murine macrophage RAW 264.7 cell line was purchased from ATCC (Manassas, VA) and maintained according to their instructions. Cells were subcultured in 96-well plates at a density of 8x 104 cells per well and allowed to reach 90% confluence, approximately 2 days. LPS (1 μg /ml) or PBS alone was added to the cell media and incubated for 12 hrs. The media was removed from the wells and LPS (1 μg/ml) with the test compounds dissolved in DMSO and serum-free RPMI, were added to the wells to achieve final concentrations of MgRIAA at 20, 5.0, 1.0 and 0.1 μg/ml and celecoxib at 100, 10, 1 and 0.1 ng/ml. Each concentration was run in 8 duplicates. Following 1 hr of incubation with the test compounds, the cell media were removed and replaced with fresh media with test compounds with LPS (1 μg/ml) and incubated for 1 hr. The media were removed from the wells and analyzed for the PGE2 synthesis.
[00187] PGE2 assay - A commercial, non-radioactive procedure for quantification of
PGE2 was employed (Cayman Chemical, Ann Arbor, MI). Samples were diluted 10 times in ElA buffer and the recommended procedure of the manufacturer was used without modification. The PGE2 concentration was represented as picograms per ml. 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 - lOOO pg ml"1.
[00188] COX-2 specific inhibitor celecoxib dose-dependently inhibited COX-2 mediated PGE2 synthesis (100, 10, 1 and 0.1 ng/ml) while no significant PGE2 inhibition was observed with MgRIAA. The data suggest that MgRIAA is not a direct COX-2 enzymatic inhibitor like celocoxib (Fig. 5)
Example 9
Inhibition of iNOS and COX-2 protein expression by MgRIAA
[00189] Cellular extracts from RAW 264.7 cells treated with MgRIAA and stimulated with LPS were assayed for iNOS and COX-2 protein by Western blot. [00190] Materials - Test compounds were prepared in dimethyl sulfoxide (DMSO) and stored at -200C. MgRIAA was supplied by Metageπics (San Clemente, CA). Parthenolide was purchased from Sigma-Aldrich (St. Louis, MO). The PI3K inhibitors wortmannin and LY294002 were purchased from EMD Biosciences (San Diego, CA). Antibodies generated against COX-2 and iNOS were purchased from Cayman Chemical (Ann Arbor, MI). Antibodies generated against GAPDH were purchased from Novus Biological (Littleton, CO). Secondary antibodies coupled to horseradish peroxidase were purchased from Amersham Biosciences (Piscataway, NJ).
[00191] Cell Culture — The murine macrophage RAW 264.7 cell line was purchased from ATCC (Manassas, VA) and maintained according to their instructions. Cells were grown and subcultured in 24- well plates at a density of 3 x 105 cells per well and allowed to reach 90% confluence, approximately 2 days. Test compounds were added to the cells in serum free medium at a final concentration of 0.4% DMSO. Following 1 hr of incubation with the test compounds, LPS (1 μg/ml) or phosphate buffered saline alone was added to the cell wells and incubation continued for the indicated times.
[00192] Western Blot - Cell extracts were prepared in Buffer E (50 mM HEPES, pH
7.0; 150 mM NaCl; 1% triton X-100; 1 mM sodium orthovanadate; aprotinin 5 μg/ml; pepstatin A 1 μg/ml; leupeptin 5 μg/ml; phenylmethanesulfonyl fluoride 1 mM). Briefly, cells were washed twice with cold PBS and Buffer E was added. Cells were scraped into a clean tube, following a centrifugation at 14,000 rpm for 10 minutes at 4°C, the supernatant was taken as total cell extract. Cell extracts (50 μg) were electrophoresed through a pre-cast 4%-20% Tris-HCl Criterion gel (Bio-Rad, Hercules, CA) until the front migration dye reached 5 mm from the bottom of the gel. The proteins were transferred to nitrocellulose membrane using a semi-dry system from Bio-Rad (Hercules, CA). The membrane was washed and blocked with 5% dried milk powder for 1 hour at room temperature. Incubation with the primary antibody followed by the secondary antibody was each for one hour at room temperature. Chemiluminescence was performed using the SuperSignal West Femto Maximum Sensitivity Substrate from Pierce Biotechnology (Rockford, IL) by incubation of equal volume of luminol/enhancer solution and stable peroxide solution for 5 minutes at room temperature. The Western blot image was captured using a cooled CCD Kodak® (Rochester, NY) ISlOOO imaging system. Densitometry was performed using Kodak® software. [00193] The percent of COX-2 and iNOS protein expression was assessed using
Western blot detection. The expression of COX-2 was observed after 20 hours stimulation with LPS. As compared to the solvent control of DMSO, a reduction of 55% was seen in COX-2 protein expression by MgRIAA (Fig. 6). A specific NF-kB inhibitor parthenolide, inhibited protein expression 22.5%, while the PI3-kinase inhibitor decreased COX-2 expression about 47% (Fig. 6). Additionally, a reduction of 73% of iNOS protein expression was observed after 20 hr stimulation with LPS (Fig. 7) by MgRIAA.
Example 10
NF-κB nuclear translocation and DNA Binding
[00194] Nuclear extracts from RAW 264.7 cells treated with MgRIAA and stimulated with LPS for 4 hours were assayed for NF-κB binding to DNA.
[00195] Materials — Test compounds were prepared in dimethyl sulfoxide (DMSO) and stored at -200C. MgRIAA was supplied by Metagenics (San Clemente, CA). Parthenolide, a specific inhibitor for NF-kB activation was purchased from Sigma-Aldrich (St. Louis, MO). The PI3K inhibitor LY294002 was purchased from EMD Biosciences (San Diego, CA).
[00196] Cell Culture - The murine macrophage RAW 264.7 cell line was purchased from ATCC (Manassas, VA) and maintained according to their instructions. Cells were subcultured in 6-well plates at a density of 1.5 x 106 cells per well and allowed to reach 90% confluence, approximately 2 days. Test compounds MgRIAA (55 and 14 μg/ml), parthenolide (80 μM) and LY294002 (25 μM) were added to the cells in serum free media at a final concentration of 0.4% DMSO. Following 1 hr of incubation with the test compounds, LPS (1 μg/ml) or PBS alone was added to the cell media and incubation continued for an additional four hours.
[00197] NF-KB-DNA binding - Nuclear extracts were prepared essentially as described by Dignam, et al [Nucl Acids Res 11:1475-1489, (1983)]. Briefly, cells were washed twice with cold PBS, then Buffer A (10 mM HEPES, pH 7.0; 1.5 mM MgCl2; 10 mM KCl; 0.1% NP-40; aprotinin 5 μg/ml; pepstatin A 1 μg/ml; leupeptin 5 μg/ml; phenylmethanesulfonyl fluoride 1 mM) was added and allowed to sit on ice for 15 minutes. Cells were then scraped into a clean tube and processed through three cycles of freeze/thaw. The supernatant layer following centrifugation at 10,000 x g for 5 min at 4°C was the cytoplasmic fraction. The remaining pellet was resuspended in Buffer C (20 mM HEPES, pH 7.0; 1.5 mM KCl; 420 mM KCl; 25% glycerol; 0.2 M EDTA; aprotinin 5 μg/ml; pepstatin A 1 μg/ml; leupeptin 5 μg/ml; phenylmethanesulfonyl fluoride 1 mM) and allowed to sit on ice for 15 minutes. The nuclear extract fraction was collected as the supernatant layer following centrifugation at 10,000 x g for 5 min at 4°C. NF-kB DNA binding of the nuclear extracts was assessed using the TransAM NF-κB kit from Active Motif (Carlsbad, CA) as per manufacturer's instructions. As seen in Figure 8, the TransAM kit detected the p50 subunit of NF-κB binding to the consensus sequence in a 96-well format. Protein concentration was measured (Bio-Rad assay) and 10 μg of nuclear protein extracts were assayed in duplicate.
[00198] Analysis of nuclear extracts (10 μg protein) was performed in duplicate and the results are presented graphically in Figure 9. Stimulation with LPS (1 μg/ml) resulted in a two-fold increase in NF-κB DNA binding. Treatment with LY294002 (a PB kinase inhibitor) resulted in a modest decrease of NF-ΛB binding as expected from previous literature reports. Parthenolide also resulted in a significant reduction in NF-κB binding as expected. A large reduction of NF-κB binding was observed with MgRIAA. The effect was observed in a dose-response manner. The reduction in NF-/cB binding may result in reduced transcriptional activation of target genes, including COX-2, iNOS and TNFα.
[00199] The results suggest that the decreased NF-κB binding observed with
MgDHIAA may result in decreased COX-2 protein expression, ultimately leading to a decrease in PGE2 production.
Example 11
Increased lipogenesis in 3T3-L1 adipocytes elicited by a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia bark.
[00200] The Model - The 3T3-L1 murine fibroblast model is used to study the potential effects of compounds on adipocyte differentiation and adipogenesis. This cell line allows investigation of stimuli and mechanisms that regulate preadipocytes replication separately from those that regulate differentiation to adipocytes [Fasshauer, M., Klein, J., Neumann, S., Eszlinger, M., and Paschke, R. Hormonal regulation of adiponectin gene expression in 3T3- Ll adipocytes. Biochem Biophys Res Commun, 290: 1084-1089, (2002); Li, Y. and Lazar, M. A. Differential gene regulation by PPARgamma agonist and constitutively active PPARgamma2. MoI Endocrinol, 16: 1040-1048, (2002)] as well as insulin-sensitizing and triglyceride-lowering ability of the test agent [Raz, L, Eldor, R., Cernea, S., and Shafhr, E. Diabetes: insulin resistance and derangements in lipid metabolism. Cure through intervention in fat transport and storage. Diabetes Metab Res Rev, 21: 3-14, (2005)].
[00201] As preadipocytes, 3T3-L1 cells have a fibroblastic appearance. They replicate in culture until they form a confluent monolayer, after which cell-cell contact triggers Go/G] growth arrest. Terminal differentiation of 3T3-L1 cells to adipocytes depends on proliferation of both pre- and post-confluent preadipocytes. Subsequent stimulation with 3- isobutyl-1-methylxanthane, dexamethasone, and high does of insulin (MDI) for two days prompts these cells to undergo post-confluent mitotic clonal expansion, exit the cell cycle, and begin to express adipocyte-specific genes. Approximately five days after induction of differentiation, more than 90% of the cells display the characteristic lipid-filled adipocyte phenotype. Assessing triglyceride synthesis of 3T3-L1 cells provides a validated model of the insulin-sensitizing ability of the test agent
[00202] It appears paradoxical that an agent that promotes lipid uptake in fat cells should improve insulin sensitivity. Several hypotheses have been proposed in an attempt to explain this contradiction. One premise that has continued to gain research support is the concept of "fatty acid steal" or the incorporation of fatty acids into the adipocyte from the plasma causing a relative depletion of fatty acids in the muscle with a concomitant improvement of glucose uptake [Martin, G., K. Schoonjans, et al. PPARgamma activators improve glucose homeostasis by stimulating fatty acid uptake in the adipocytes. Atherosclerosis 137 Suppl: S75-80, (1998)]. Thiazolidinediones, such as troglitazone and pioglitazone, have been shown to selectively stimulate lipogenic activities in fat cells resulting in greater insulin suppression of lipolysis or release of fatty acids into the plasma [Yamauchi, T., J. Kamon, et al. The mechanisms by which both heterozygous peroxisome proliferator-activated receptor gamma (PPARgamma) deficiency and PPARgamma agonist improve insulin resistance. J Biol Chem 276(44): 41245-54, (2001); Oakes, N. D., P. G. Thalen, et al. Thiazolidinediones increase plasma-adipose tissue FFA exchange capacity and enhance insulin-mediated control of systemic FFA availability. Diabetes 50(5): 1158-65, (2001)]. This action would leave less free fatty acids available for other tissues [Yang, W. S., W. J. Lee, et al. Weight reduction increases plasma levels of an adipose-derived antiinflammatory protein, adiponectin. J Clin Endocrinol Metab 86(8): 3815-9, (2001)]. Thus, insulin desensitizing effects of free fatty acids in muscle and liver would be reduced as a consequence of thiazolidinedione treatment. These in vitro results have been confirmed clinically [Boden, G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 46(1): 3-10, (1997); Stumvoll, M. and H. U. Haring Glitazones: clinical effects and molecular mechanisms. Ann Med 34(3): 217-24, (2002)].
(00203] Test Materials — Troglitazone was obtained from Cayman Chemicals (Ann
Arbor, Ml, while methylisobutylxanthine, dexamethasone, indomethacin, Oil red O and insulin were obtained from Sigma (St. Louis, MO). The test material was a dark brown powder produced from a 50:50 (v/v) water/alcohol extract of the gum resin of Acacia (AcE) sample #4909 and was obtained from Bayir Chemicals (No. 68, South Cross Road, Basavanagudi, India). The extract was standardized to contain not less than 20% apecatechin. Batch No. A Cat/2304 used in this example contained 20.8% apecatechin as determined by UV analysis. Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS-HI (fetal bovine serum-heat inactivated) from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
[002041 Cell culture and Treatment - The murine fibroblast cell line 3T3-L1 was purchased from the American Type Culture Collection (Manassas, VA) and sub-cultured according to instructions from the supplier. Prior to experiments, cells were cultured in DMEM containing 10% FBS-HI added 50 units penicillin/ml and 50 μg streptomycin/ml, and maintained in log phase prior to experimental setup. Cells were grown in a 5% CO2 humidified incubator at 37°C. Components of the pre-confluent medium included (1) 10% FBS/DMEM containing 4.5 g glucose/L; (2) 50 U/ml penicillin; and (3) 50 μg/ml streptomycin. Growth medium was made by adding 50 ml of heat inactivated FBS and 5 ml of penicillin/streptomycin to 500 ml DMEM. This medium was stored at 4°C. Before use, the medium was warmed to 37°C in a water bath.
[00205] 3T3-TI cells were seeded at an initial density of 6x104 cells/cm2 in 24-well plates. For two days, the cells were allowed grow to reach confluence. Following confluence, the cells were forced to differentiate into adipocytes by the addition of differentiation medium; this medium consisted of (1) 10% FBS/DMEM (high glucose); (2) 0.5 mM methylisobutylxanthine; (3) 0.5 μM dexamethasone and (4) 10 μg/ml insulin (MDI medium). After three days, the medium was changed to post-differentiation medium consisting of 10 μg/ml insulin in 10% FBS/DMEM.
[00206] AcE was partially dissolved in dimethyl sulfoxide (DMSO) and added to the culture medium to achieve a concentration of 50 μg/ml at Day 0 of differentiation and throughout the maturation phase (Days 6 or 7 (D6/7)). Whenever fresh media were added, fresh test material was also added. DMSO was chosen for its polarity and the fact that it is miscible with the aqueous cell culture media. As positive controls, indomethacin and troglitazone were added, respectively, to achieve final concentrations of 5.0 and 4.4 μg/ml. Differentiated, D6/D7 3T3-L1 cells were stained with 0.36% Oil Red O or 0.001% BODIPY. The complete procedure for differentiation and treatment of cells with test materials is outlined schematically in Figure 10.
[002071 Oil Red O Staining - Triglyceride content of D6/D7-differentiated 3T3-L1 cells was estimated with Oil Red O according to the method of Kasturi and Joshi [Kasturi, R. and Joshi, V. C. Hormonal regulation of stearoyl coenzyme A desaturase activity and lipogenesis during adipose conversion of 3T3-L1 cells. J Biol Chem, 257: 12224-12230, 1982]. Monolayer cells were washed with PBS (phosphate buffered saline, Mediatech) and fixed with 10% formaldehyde for ten minutes. Fixed cells were stained with an Oil Red O working solution of three parts 0.6% Oil Red O/isopropanol stock solution and two parts water for one hour and the excess stain was washed once with water. The resulting stained oil droplets were extracted from the cells with isopropanol and quantified by spectrophotometric analysis at 540 nm (MEL312e BIO-KINETICS READER, Bio-Tek Instruments, Winooski, VT). Results for test materials and the positive controls indomethacin and troglitazone were represented relative to the 540 nm absorbance of the solvent controls.
[00208] BODIPY Staining - 4,4-Difluoro-l ,3,5,7,8-penta-methyl-4-bora-3a,4a-diaza-s- indacene (BODIPY 493/503; Molecular Probes, Eugene, OR) was used for quantification of cellular neutral and nonpolar lipids. Briefly, media were removed and cells were washed once with non-sterile PBS. A stock IOOOX BODIPY/DMSO solution was made by dissolving 1 mg BODIPY in 1 ml DMSO (1,000 μg BODIPY/ml). A working BODEPY solution was then made by adding 10 μl of the stock solution to 990 μl PBS for a final BODIP Y concentration in the working solution of 0.01 μg/μl. One-hundred μl of this working solution (1 μg BODIPY) was added to each well of a 96-well microtiter plate. After 15 min on an orbital shaker (DS-500, VWR Scientific Products, South Plainfield, NJ) at ambient temperature, the cells were washed with 100 μl PBS followed by the addition of 100 μl PBS for reading for spectrofluorometric determination of BODIPY incorporation into the cells. A Packard Fluorocount spectrofluorometer (Model#BF 10000, Meridan, CT) set at 485 nm excitation and 530 nm emission was used for quantification of BODIPY fluorescence. Results for test materials, indomethacin, and troglitazone were reported relative to the fluorescence of the solvent controls.
[00209J A chi-square analysis of the relationship between the BODEPY quantification of all neutral and nonpolar lipids and the Oil Red O determination of triglyceride content in 3T3-L1 cells on D7 indicated a significant relationship between the two methods with pO.OOl and Odds Ratio of 4.64.
[00210] Statistical Calculations and Interpretation - AcE and indomethacin were assayed a minimum of three times in duplicate. Solvent and troglitazone controls were replicated eight times also in duplicate. Nonpolar lipid incorporation was represented relative to the nonpolar lipid accumulation of fully differentiated cells in the solvent controls. A positive response was defined as an increase in lipid accumulation assessed by Oil Red O or BODIPY staining greater than the respective upper 95% confidence interval of the solvent control (one-tail, Excel; Microsoft, Redmond, WA). AcE was further characterized as increasing adipogenesis better than or equal to the troglitazone positive control relative to the solvent response; the student t-test function of Excel was used for this evaluation.
[00211] Results — The positive controls indomethacin and troglitazone induced lipogenesis to a similar extent in 3T3-L1 cells (Figure 11). Unexpectedly, the AcE produced an adipogenic response greater than either of the positive controls indomethacin and troglitazone.
[00212] The lipogenic potential demonstrated in 3T3-L1 cells, dimethyl sulfoxide- soluble components of an aqueous Acacia sample #4909 extract demonstrates a potential to increase insulin sensitivity in humans or other animals exhibiting signs or symptoms of insensitivity to insulin.
Example 12 Increased adiponectin secretion from insulin-resistant 3T3-L1 adipocytes elicited by a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia.
[00213] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments.
[00214J Test Materials — Troglitazone was purchased from Cayman Chemical (Ann
Arbor, MI) while methylisobutylxanthine, dexamethasone, and insulin were obtained from Sigma (St. Louis, MO). The test material was a dark brown powder produced from a 50:50 (v/v) water/alcohol extract of the gum resin of Acacia sample #4909 and was obtained from Bayir Chemicals (No. 68, South Cross Road, Basavanagudi, India). The extract was standardized to contain not less than 20% apecatechin. Batch No. A Cat/2304 used in this example contained 20.8% apecatechin as determined by UV analysis. Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS-HI (fetal bovine serum-heat inactivated from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
[00215] Cell culture and Treatment - Culture of the murine fibroblast cell line 3T3-L1 to produce Day 6 differentiated adipocytes was performed as described in Example 10. 3T3- Ll cells were seeded at an initial density of 1x104 cells/cm2 in 96-well plates. For two days, the cells were allowed grow to reach confluence. Following confluence, the cells were forced to differentiate into adipocytes by the addition of differentiation medium; this medium consisted of (1) 10% FBS/DMEM (high glucose); (2) 0.5 mM methylisobutylxanthine; (3) 0.5 μM dexamethasone and (4) 10 μg/ml insulin (MDI medium). From Day 3 through Day 5, the medium was changed to post-differentiation medium consisting of 10 μg/ml insulin in 10% FBS/DMEM.
[00216] Assessing the effect of Acacia on insulin-resistant, mature 3T3-L1 cells was performed using a modification of the procedure described by Fasshauer et al. [Fasshauer, et al. Hormonal regulation of adiponectin gene expression in 3T3-L1 adipocytes. BBRC 290:1084-1089, (2002)]. Briefly, on Day 6, cells were maintained in serum-free media containing 0.5% bovine serum albumin (BSA) for three hours and then treated with 1 μg insulin/ml plus solvent or insulin plus test material. Troglitazone was dissolved in dimethyl sulfoxide and added to achieve concentrations of 5, 2.5, 1.25 and 0.625 μg/ml. The Acacia extract was tested at 50, 25, 12.5 and 6.25 μg/ml. Twenty-four hours later, the supernatant medium was sampled for adiponectin determination. The complete procedure for differentiation and treatment of cells with test materials is outlined schematically in Figure 12.
[00217] Adiponectin Assay — The adiponectin secreted into the medium was quantified using the Mouse Adiponectin Quantikiηe® Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN)- Information supplied by the manufacturer indicated that recovery of adiponectin spiked in mouse cell culture media averaged 103% and the minimum detectable adiponectin concentration ranged from 0.001 to 0.007 ng/ml.
[00218] Statistical Calculations and Interpretation — All assays were preformed in duplicate. For statistical analysis, the effect of Acacia on adiponectin secretion was computed relative to the solvent control. Differences between the doses were determined using the student's t-test without correction for multiple comparisons; the nominal five percent probability of a type I error was selected.
[00219] Potency of the test materials was estimated using a modification of the method of Hofstee [Hofstee, B.H. Non-inverted versus inverted plots in enzyme kinetics. Nature 184:1296-1298, (1959)] for determination of the apparent Michaelis constants and maximum velocities. Substituting {relative adiponectin secretion/[concentration]} for the independent variable v/[S] and {relative adiponectin secretion} for the dependant variable {v}5 produced a relationship of the form y = mx + b. Maximum adiponectin secretion relative to the solvent control was estimated from the y-intercept, while the concentration of test material necessary for half maximal adiponectin secretion was computed from the negative value of the slope.
[00220] Results - All concentrations tested for the positive control troglitazone enhanced adiponectin secretion with maximal stimulation of 2.44-fold at 2.5 μg/ml relative to the solvent control in insulin-resistant 3T3-L1 cells (Figure 13). Both the 50 and 25 μg Acacia/ml concentrations increased adiponectin secretion relative to the solvent controls 1.76- and 1.70-fold respectively. While neither of these concentrations of Acacia was equal to the maximal adiponectin secretion observed with troglitazone, they were comparable to the 1.25 and 0.625 μg/ml concentrations of troglitazone.
[00221] Estimates of maximal adiponectin secretion derived from modified Hofstee plots indicated a comparable relative increase in adiponectin secretion with a large difference in concentrations required for half maximal stimulation. Maximum adiponectin secretion estimated from the y-intercept for troglitazone and Acacia catechu was, respectively, 2.29- and 1.88-fold relative to the solvent control. However, the concentration required for stimulation of half maximal adiponectin secretion in insulin-resistant 3T3-L1 cells was 0.085 μg/ml for troglitazone and 5.38 μg/ml for Acacia. Computed upon minimum apecatechin content of 20%, this latter figure for Acacia becomes approximately 1.0 μg/ml.
(00222] Based upon its ability to enhance adiponectin secretion in insulin-resistant
3T3-L1 cells, Acacia, and/or apecatechin, may be expected to have a positive effect on clinical pathologies in which plasma adiponectin concentrations are depressed.
Example 13
Increased adiponectin secretion from TNFα-treated 3T3-L1 adipocytes elicited by a dimethyl sulfoxi de-soluble fraction of an aqueous extract of Acacia.
[00223] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments.
[00224] Test Materials - Indomethacin, methylisobutylxanthine, dexamethasone, and insulin were obtained from Sigma (St. Louis, MO). The test material was a dark brown powder produced from a 50:50 (v/v) water/alcohol extract of the gum resin of Acacia sample #4909 and was obtained from Bayir Chemicals (No. 68, South Cross Road, Basavanagudi, India). The extract was standardized to contain not less than 20% apecatechin. Batch No. A Cat/2304 used in this example contained 20.8% apecatechin as determined by UV analysis. Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS (fetal bovine serum) characterized from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
[00225] Cell culture and Treatment - Culture of the murine fibroblast cell line 3T3-L1 to produce Day 3 differentiated adipocytes was performed as described in Example 10. 3T3- Ll cells were seeded at an initial density of IxIO4 cells/cm2 in 96-well plates. For two days, the cells were allowed grow to reach confluence. Following confluence, the cells were forced to differentiate into adipocytes by the addition of differentiation medium; this medium consisted of (1) 10% FBS/DMEM (high glucose); (2) 0.5 mM methylisobutylxanthine; (3) 0.5 μM dexamethasone and (4) 10 μg/ml insulin (MDI medium). From Day 3 through Day 5, the medium was changed to post-differentiation medium consisting of 10% FBS in DMEM. On Day 5 the medium was changed to test medium containing 10, 2 or 0.5 ng TNFo/ml in 10% FBS/DMEM with or without indomethacin or Acacia extract. Indomethacin was dissolved in dimethyl sulfoxide and added to achieve concentrations of 5, 2.5, 1.25 and 0.625 μg/ml. The Acacia extract was tested at 50, 25, 12.5 and 6.25 μg/ml. On Day 6, the supernatant medium was sampled for adiponectin determination. The complete procedure for differentiation and treatment of cells with test materials is outlined schematically in Figure 14.
[00226] Adiponectin Assay — The adiponectin secreted into the medium was quantified using the Mouse Adiponectin Quantikine® Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN). Information supplied by the manufacturer indicated that recovery of adiponectin spiked in mouse cell culture media averaged 103% and the minimum detectable adiponectin concentration ranged from 0.001 to 0.007 ng/ml.
Ϊ00227] Statistical Calculations and Interpretation — All assays were preformed in duplicate. For statistical analysis, the effect of indomethacin or Acacia catechu on adiponectin secretion was computed relative to the solvent control. Differences among the doses and test agents were determined using the Student's t-test without correction for multiple comparisons; the nominal five percent probability of a type I error was selected.
[00228] Results - TNFa significantly (p<0.05) depressed adiponectin secretion 65 and
29%, respectively, relative to the solvent controls in mature 3T3-L1 cells at the 10 and 2 ng/ml concentrations and had no apparent effect on adiponectin secretion at 0.5 ng/ml (Figure 15). At 10 and 2 ng TNFaZmI, indomethacin enhanced (p<0.05) adiponectin secretion relative to TNFor alone at all doses tested, but failed to restore adiponectin secretion to the level of the solvent control. Acacia treatment in the presence of 10 ng TNFoΛnl, produced a similar, albeit attenuated, adiponectin increase relative to that of indomethacin. The differences in adiponectin stimulation between Acacia catechu and indomethacin were 14, 20, 32, and 41%, respectively, over the four increasing doses. Since the multiple between doses was the same for indomethacin and Acacia, these results suggest that the potency of indomethacin was greater than the active material(s) in Acacia at restoring adiponectin secretion to 3T3-L1 cells in the presence of supraphysiological concentrations of TNFα. [00229] Treatment of 3T3-L1 ceils with 2 ng TNFα and Acacia produced increases in adiponectin secretion relative to TNFa alone that were significant (p<0.05) at 6.25, 25 and 50 μg/ml. Unlike the 10 ng TNFo/ml treatments, however, the differences between Acacia and indomethacin were smaller and not apparently related to dose, averaging 5.5% over all four concentrations tested. As observed with indomethacin, Acacia did not restore adiponectin secretion to the levels observed in the solvent control.
[00230] At 0.5 ng TNFαΛnl, indomethacin produced a dose-dependant decrease in adiponectin secretion that was significant (p<0.05) at the 2.5 and 5.0 μg/ml concentrations. Interestingly, unlike indomethacin, Acacia catechu increased adiponectin secretion relative to both the TNFαr and solvent treated 3T3-L1 adipocytes at 50 μg/ml. Thus, at concentrations of TNFα approaching physiologic levels, Acacia catechu enhanced adiponectin secretion relative to both TNFα and the solvent controls and, surprisingly, was superior to indomethacin.
[00231] Based upon its ability to enhance adiponectin secretion in TNFα-treated 3T3-
Ll cells, Acacia catechu, and/or apecatechin, would be expected to have a positive effect on all clinical pathologies in which TNFα levels are elevated and plasma adiponectin concentrations are depressed.
Example 14
A variety of commercial Acacia samples increase lipogenesis in the 3T3-L1 adipocyte model.
[00232] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. All chemicals and procedures used were as described in Example 11 with the exception that only the Oil Red O assay was performed to assess Acacia catechu-induced, cellular triglyceride content. Acacia catechu sample #5669 was obtained from Natural Remedies (364, 2nd Floor, 16th Main, 4th T Block Bangalore, Karnataka 560041 India); and samples #4909, #5667, and #5668 were obtained from Bayir Chemicals (No. 10, Doddanna Industrial Estate, Penya II Stage, Bangalore, 560091 Karnataka, India). Acacia nilotica samples #5639, #5640 and #5659 were purchased from KDN- Vita International, Inc. (121 Stryker Lane, Units 4 & 6 Hillsborough, NJ 08844). Sample #5640 was described as bark, sample #5667 as a gum resin and sample #5669 as heartwood powder. All other samples unless indicated were described as proprietary methanol extracts of Acacia catechu bark.
[00233] Results — All Acacia samples examined produced a positive lipogenic response
(Figure 16). The highest lipogenic responses were achieved from samples #5669 the heartwood powder (1.27), #5659 a methanol extract (1.31), #5640 a DMSO extract (1.29) and #4909 a methanol extract (1.31).
[00234] This example further demonstrates the presence of multiple compounds in
Acacia catechu that are capable of positive modification of adipocyte physiology supporting increased insulin actions.
Example 15
A variety of commercial Acacia samples increase adiponectin secretion the TNFo:-3T3-Ll adipocyte model.
[00235] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals used and treatment of cells was performed as noted in Examples 11 and 13. Treatment of 3T3-L1 adipocytes with TNFα differed from Example 12, however, in that cells were exposed to 2 or 10 ng TNFα/rnl only. On Day 6 culture supernatant media were assayed for adiponectin as detailed in Example 12. Formulations of Acacia samples #4909, #5639, #5659, #5667, #5668, #5640, and #5669 were as described in Example 13.
[00236] Results - The 2 ng/ml TNFa reduced adiponectin secretion of 3T3-L1 adipocytes by 27% from the solvent control, while adiponectin secretion was maximally elevated 11% from the TNFα solvent control by 1.25 μg indomethacin/ml (Table 12). Only Acacia formulation #5559 failed to increase adiponectin secretion at any of the four doses tested. All other formulations of Acacia produced a comparable maximum increase of adiponectin secretion ranging from 10 to 15%. Differences were observed, however, with regard to the concentrations at which maximum adiponectin secretion was elicited by the various Acacia formulations. The most potent formulation was #5640 with a maximal stimulation of adiponectin stimulation achieved at 12.5 μg/ml, followed by #4909 and #5668 at 25 μg/ml and finally #5639, #5667 and #5669 at 50 μg/ml. Table 12
Relative maximum adiponectin secretion from 3T3-L1 adipocytes elicited by various formulations of Acacia in the presence of 2 ng TNFo/ml.
Figure imgf000073_0001
t Adiponectin Index = [Adiponectin]τest/[Adiponectin]τNFα control *Significantly increased (p<0.05) from TNFα solvent response.
[00237] The 10 ng/ml TNFα reduced adiponectin secretion of 3T3-L1 adipocytes by
54% from the solvent control, while adiponectin secretion was maximally elevated 67% from the TNFαr solvent control by 5.0 μg indomethacin/ml (Table 13). Troglitazone maximally increased adiponectin secretion 51% at the lowest dose tested 0.625 μg/ml. Acacia formulation #5559 produced the lowest significant increase (p<0.05) of 12% at 25 μg/ml. All other formulations of Acacia produced a maximum increase of adiponectin secretion at 50 μg/ml ranging from 17 to 41%. The most potent formulations were #4909 and #5669 with increases in adiponectin secretion of 41 and 40%, respectively over the TNFα solvent control.
Table 13
Relative maximum adiponectin secretion from 3T3-L1 adipocytes elicited by various formulations of Acacia in the presence of 10 n ε TNFo^mI.
Figure imgf000073_0002
Figure imgf000074_0001
t Adiponectin Index = [Adiponectin]τest/[Adiponectin]τNFα control *Significantly increased (p<0.05) from TNFα solvent response.
[00238] The observation that different samples or formulations of Acacia elicit similar responses in this second model of metabolic syndrome, further demonstrates the presence of multiple compounds in Acacia that are capable of positive modification of adipocyte physiology supporting increased insulin actions.
Example 16
Polar and non-polar solvents extract compounds from Acacia catechu capable of increasing adiponectin secretion in the TNPo/3T3-Ll adipocyte model.
[00239] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals used are as noted in Examples 11 and 13. 3T3-L1 adipocytes were treated with 10 ng TNFαΛnl as described in Example 13. Culture supernatant media were assayed for adiponectin on Day 6 as detailed in Example 13.
[00240] Test Materials - Large chips of Acacia catechu sample #5669 heartwood (each chip weighing between 5-10 grams) were subjected to drilling with a 5/8" metal drill bit using a standard power drill at low speed. The wood shavings were collected into a mortar, and ground into a fine powder while frozen under liquid N2. This powder was then sieved through a 250 micron screen to render approximately 10 g of a fine free-flowing powder.
Table 14
Description of Acacia catechu extraction samples for 3T3-L1 adiponectin assay.
Figure imgf000075_0001
Gastric fluid consisted of 2.90 g NaCl, 7.0 m concentrated, aqueous HCl, 3.2 g pepsin (800 - 2500 activity units/mg) diluted to 1000 ml with water. Final pH was 1.2. For this extraction, the gastric fluid-heartwood suspension remained at 40° C for one hour followed by removal of the gastric fluid in vacuo. The remaining residue was then dissolved in MeOH, filtered through a 0.45 micron PTFE syringe filter and concentrated in vacuo.
(00241) This powder was dispensed into six glass amber vials (150 mg/vial) and extracted at 400C for approximately 10 hr with 2 ml of the solvents listed in Table 14. Following this extraction, the heartwood/solvent suspensions were subjected to centrifugation (5800 x g, 10 min.). The supernatant fractions from centrifugation were filtered through a 0.45 micron PTFE syringe filter into separate amber glass vials. Each of these samples was concentrated in vacuo. As seen in Table 7, DMSO extracted the most material from the Acacia catechu heartwood and chloroform extracted the least. All extract samples were tested at 50, 25, 12.5, and 6.25 μg/ml.
[00242] Pioglitazone was obtained as 45 mg pioglitazone tables from a commercial source as Actos® (Takeda Pharmaceuticals, Lincolnshire, IL). The tablets were ground to a fine powder and tested at 5.0, 2.5, 1.25 and 0.625 μg pioglitazone/ml. Indomethacin was also included as an additional positive control.
100243] Results — Both positive controls pioglitazone and indomethacin increased adiponectin secretion by adipocytes in the presence of TNFα; 115 and 94% respectively (Figure 17). Optimal pioglitazone and indomethacin concentrations were, 1.25 and 2.5 μg/ml respectively. All extracts of Acacia catechu sample #5669 increased adiponectin secretion relative to the TNFa treatment. Among the extracts, the DMSO extract was the most potent inducer of adiponectin secretion with maximal activity observed at 6.25 μg extract/ml. This result may be due to the ability of DMSO to extract a wide range of materials of varying polarity. An examination of Figure 17 indicates that both the water extract (polar compounds) and the chloroform extract (nonpolar compounds) were similar in their ability to increase adiponectin secretion in the TNFo/3T3-Ll adipocyte model. It is unlikely that these extracts contained similar compounds. This example illustrates the ability of solvents with differing polarities to extract compounds from Acacia catechu heartwood that are capable of increasing adiponectin secretion from adipocytes in the presence of a pro-inflammatory stimulus.
Example 17
Acacia catechu acidic and basic fractions are capable of increasing adiponectin secretion in the TNFQ/3T3-L1 adipocyte model.
[00244] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals used were as noted in Examples 11 and 13. 3T3-L1 adipocytes were treated with 10 ng TNFα^ml as described in Example 13. Culture supernatant media were assayed for adiponectin on Day 6 as detailed in Example 13.
[00245] Test Materials — Acacia catechu sample #5669 was extracted according to the following procedure: Alkaline isopropyl alcohol solution, (1% (v/v) 1.5N NaOH in isopropanol,) was added to approximately 50 mg of the dry Acacia catechu heartwood powder #5669 in a 50 ml tube. The sample was then mixed briefly, sonicated for 30 minutes, and centrifuged for an hour to pellet the remaining solid material. The supernatant liquid was then filtered through 0.45 micron filter paper. The pH of the basic isopropanol used was pH 8.0, while the pH of the collected liquid was pH 7.0. A portion of the clear, filtered liquid was taken to dryness in vacuo and appeared as a white solid. This sample was termed the dried alkaline extract.
[00246] The remaining pelleted material was brought up in acidic isopropyl alcohol solution, (1% (v/v) 10% HCl in isopropanol,) as a red solution. This sample was mixed until the pellet material was sufficiently dispersed in the liquid and then centrifuged for 30 minutes to again pellet the remaining solid. The pale yellow supernatant fluid was passed through a 0.45 micron filter paper. The pH of the collected liquid was pH 3.0 and it was found that in raising the pH of the sample to pH 8-9 a reddish-brown precipitate was formed (dried precipitate). The precipitate was collected and dried, providing a reddish-brown solid. The supernatant liquid was again passed through a 0.45 micron filter to remove any remaining precipitate; this liquid was a deep yellow color. This remaining liquid was taken to dryness resulting in a solid brown sample and termed dried acidic extract. Recoveries for the three factions are listed in Table 15. All test materials were assayed at 50, 25, 12.5 and 6.25 μg/ml, while the pioglitazone positive control was tested at 5.0, 2.5, 1.25 and 0.625 μg/ml.
Table 15
Test material recovery from Acacia catechu heartwood powder.
Figure imgf000077_0001
[00247] Results: TNFα reduced adiponectin secretion by 46% relative to the solvent control. Maximal restoration of adiponectin secretion by pioglitazone was 1.47 times the TNFα treatment observed at 1.25 μg/ml (Table 16). Of the test materials, only the dried precipitant failed to increase adiponectin secretion significantly above the TNFα only control. The acidic extract and heartwood powder (starting material) were similar in their ability to increase adiponectin secretion in the presence of TNFα, while the alkaline extract increased adiponectin secretion only at the highest dose of 50 μg/ml.
Table 16
Maximum adi onectin secretion elicited over four doses in TNFα/3T3-Ll model.
Figure imgf000077_0002
ft Values > 1.1 1 are significantly different (p<0.05) from TNFα control.
Example 18
Decreased interleukin-6 secretion from TNFα-treated 3T3-L1 adipocytes by a dimethyl sulfoxide-soluble fraction of an aqueous extract of Acacia. [00248] Interleukin-6 (IL-6) is a multifunctional cytokine that plays important roles in host defense, acute phase reactions, immune responses, nerve cell functions, hematopoiesis and metabolic syndrome. It is expressed by a variety of normal and transformed lymphoid and nonlymphoid cells such as adipocytes. The production of IL-6 is up-regulated by numerous signals such as mitogenic or antigenic stimulation, lipopolysaccharides, calcium ionophores, cytokines and viruses [Hibi, M., Nakajima, K., Hirano T. IL-6 cytokine family and signal transduction: a model of the cytokine system. J MoI Med. 74(1):1-12, (Jan 1996)]. Elevated serum levels have been observed in a number of pathological conditions including bacterial and viral infection, trauma, autoimmune diseases, malignancies and metabolic syndrome [Arner, P. Insulin resistance in type 2 diabetes — role of the adipokines. Curr MoI Med.;5(3):333-9, (May 2005)].
[00249] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals used were as noted in Examples 11 and 13. 3T3-L1 adipocytes were treated with 10 ng TNFo/ml as described in Example 13. Culture supernatant media were assayed for adiponectin on Day 6 as detailed in Example 13.
[00250] Test Materials — Indomethacin, methylisobutylxanthine, dexamethasone, and insulin were obtained from Sigma (St. Louis, MO). The test material was a dark brown powder produced from a 50:50 (v/v) water/alcohol extract of the gum resin of Acacia catechu sample #4909 and was obtained from Bayir Chemicals (No. 68, South Cross Road, Basavanagudi, India). The extract was standardized to contain not less than 20% apecatechin. Batch No. A Cat/2304 used in this example contained 20.8% apecatechin as determined by UV analysis. Penicillin, streptomycin, Dulbecco's modified Eagle's medium (DMEM) was from Mediatech (Herndon, VA) and 10% FBS (fetal bovine serum) characterized from Mediatech and Hyclone (Logan, UT). All other standard reagents, unless otherwise indicted, were purchased from Sigma.
[00251] Interleukin-6 Assay — The IL-6 secreted into the medium was quantified using the Quantikine® Mouse IL-6 Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN). Information supplied by the manufacturer indicated that recovery of IL-6 spiked in mouse cell culture media averaged 99% with a 1 :2 dilution and the minimum detectable IL-6 concentration ranged from 1.3 to 1.8 pg/ml. All supernatant media samples were assayed undiluted. [00252] Statistical Calculations and Interpretation - All assays were preformed in duplicate. For statistical analysis, the effect of Acacia on adiponectin or IL-6 secretion was computed relative to the solvent control. Differences among the doses were determined using the student's t-test without correction for multiple comparisons; the nominal five percent probability of a type I error (one-tail) was selected.
[00253] Results - As seen in previous examples, TNFα dramatically reduced adiponectin secretion, while both indomethacin and the Acacia catechu extract increased adiponectin secretion in the presence of TNFα. Although both the indomethacin positive control and Acacia catechu extract demonstrated dose-related increases in adiponectin secretion, neither material restored adiponectin concentrations to those seen in the dimethyl sulfoxide controls with no TNFα (Table 17). The Acacia catechu extract demonstrated a potent, dose-related inhibition of IL-6 secretion in the presence of TNFα, whereas indomethacin demonstrated no anti-inflammatory effect.
[00254] An examination of the ratio of the anti-inflammatory adiponectin to the proinflammatory IL-6 resulted in an excellent dose-related increase in relative anti-inflammatory activity for both indomethacin and the Acacia catechu extract.
Table 17
Decreased IL-6 and increased adiponectin secretion elicited by Acacia catechu sample #4909 in the TNFoV3T3-Ll model.
Figure imgf000079_0001
The Acacia catechu test material or indomethacin was added in concert with 10 πg TNFo/ml to D5 3T3-L1 adipocytes. On the following day, supernatant media were sampled for adiponectin and IL-6 determination. All values were indexed to the TNFα control, f Adiponectin Index = [Adiponectin]τest/[Adiponectin]τNFα control ffIL-6 Index = [IL-6Test - IL-6Control]/[IL-6τNFα- IL-6Control]
Significantly different from TNFa control p<0.05).
[00255] Acacia catechu sample #4909 demonstrated a dual anti-inflammatory action in the TNFα/3T3-Ll adipocyte model. Components of the Acacia catechu extract increased adiponectin secretion while decreasing IL-6 secretion. The overall effect of Acacia catechu was strongly anti-inflammatory relative to the TNFα controls. These results support the use of Acacia catechu for modification of adipocyte physiology to decrease insulin resistance weight gain, obesity, cardiovascular disease and cancer.
Example 19
Effect of a dimethyl sulfoxide-soluble fraction of an aqueous Acacia extract on secretion of adiponectin, IL-6 and resistin from insulin-resistant 3T3-L1 adipocytes.
[002561 The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals and statistical procedures used were as noted in Examples 11 and 12. 11-6 was assayed as described in Example 18.
[00257] Resistin Assay - The amount of resistin secreted into the medium was quantified using the Quantikine® Mouse Resistin Immunoassay kit with no modifications (R&D Systems, Minneapolis, MN). Information supplied by the manufacturer indicated that recovery of resistin spiked in mouse cell culture media averaged 99% with a 1:2 dilution and the minimum detectable resistin concentration ranged from 1.3 to 1.8 pg/ml. All supernatant media samples were diluted 1 :20 with dilution media supplied by the manufacturer before assay.
[00258] Statistical Calculations and Interpretation — All assays were preformed in duplicate. For statistical analysis, the effect of Acacia catechu on adiponectin or IL-6 secretion was computed relative to the solvent control. Differences among the doses were determined using the Student's t-test without correction for multiple comparisons; the nominal five percent probability of a type I error (one-tail) was selected. W
[00259] Results - Both troglitazone and the Acacia sample #4909 increased adiponectin secretion in a dose-related manner in the presence of high concentrations of insulin (Table 18). While Acacia catechu exhibited an anti-inflammatory effect through the reduction of IL-6 at only the 6.25 μg/ml, concentration, troglitazone was pro-inflammatory at the 5.00 and 1.25 μg/ml concentrations, with no effect observed at the other two concentrations. Resistin secretion was increased in a dose-dependent fashion by troglitazone; however, Acacia catechu decreased resistin expression likewise in a dose-dependent manner.
[00260] As seen in Example 18, Acacia catechu sample #4909 again demonstrated a dual anti-inflammatory action in the hyperinsulemia/3T3-Ll adipocyte model. Components of the Acacia catechu extract increased adiponectin secretion while decreasing IL-6 secretion. Thus, the overall effect of Acacia catechu was anti-inflammatory relative to the high insulin controls. The effect of Acacia catechu on resistin secretion in the presence of high insulin concentrations was contrary to those of troglitazone: troglitazone increased resistin expression, while Acacia catechu further decreased resistin expression. These data suggest that the complex Acacia catechu extract are not functioning through PP ARγ receptors. These results provide further support the use of Acacia catechu for modification of adipocyte physiology to decrease insulin resistance weight gain, obesity, cardiovascular disease and cancer.
Table 18
Effect of Acacia catechu extract on adiponectin, IL-6 and resistin secretion in the insulin resistant 3T3-L1 model.
Figure imgf000081_0001
The Acacia catechu test material or indomethacin was added in concert with 166 nM insulin to D5 3T3-L1 adipocytes. On the following day, supernatant media were sampled for adiponectin, IL-6 and resistin determination. All values were indexed to the insulin only control. f Adiponectin Index = [Adiponectin]τest/[Adiponectin]lnsu|jn control ttIL-6 Index = [IL-6τest]/[π.-6|nsuijn control] f ftResistin Index = [Resistinτest]/[Resistininsuiin control]
*Index values represent the mean ± 95% confidence interval computed from residual mean square of the analysis of variance. Values greater or less than Insulin control ± 95% CI are significantly different with p<0.05.
Example 20
Increased lipogenesis in adipocytes by phvtochemicals derived from hops.
[00261] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals and statistical procedures used were as noted in Example 11.
[00262] Test Materials - The hops phytochemicals used in this testing are described in
Table 19 and were acquired from Betatech Hops Products (Washington, D. C, U.S.A.).
Table 19
Description of hops test materials.
Figure imgf000082_0001
Figure imgf000083_0001
[00263] Cell Culture and Treatment - Hops compounds were dissolved in dimethyl sulfoxide (DMSO) and added to achieve concentrations of 10, 5, 4 or 2 μg/ml at Day 0 of differentiation and maintained throughout the maturation phase (Days 6 or 7). Spent hops was tested at 50 μg/ml. Whenever fresh media were added, fresh test material was also added. DMSO was chosen for its polarity and the fact that it is miscible with the aqueous cell culture media. As positive controls, indomethacin and troglitazone were added, respectively, to achieve final concentrations of 5.0 and 4.4 μg/ml. Differentiated, D6/D7 3T3-L1 cells were stained with 0.36% Oil Red O or 0.001% BODIPY.
[00264] Results — The positive controls indomethacin and troglitazone induced
Iipogenesis to a similar extent in 3T3-L1 cells (Figure 18). Unexpectedly, four of the hops genera produced an adipogenic response in 3T3-L1 adipocytes greater than the positive controls indomethacin and troglitazone. These four genera included isoalpha acids, Rho- isoalpha acids, tetrahydroisoalpha acids, and hexahydroisoalpha acids. This finding is surprising in light of the published report that the binding of individual isohumulones with PPARγ was approximately one-third to one-fourth that of the potent PPAR7 agonist pioglitazone [Yajima, H., Hceshima, E., Shiraki, M., Kanaya, T., Fujiwara, D., Odai, H., Tsuboyama-Kasaoka, N., Ezaki, O., Oikawa, S., and Kondo, K. Isohumulones, bitter acids derived from hops, activate both peroxisome proliferator-activated receptor alpha and gamma and reduce insulin resistance. J Biol Chem, 279: 33456-33462, (2004)]. [00265] The adipogenic responses of xanthohumols, alpha acids and beta acids were comparable to indomethacin and troglitazone, while spent hops and hexahydrocolupulone failed to elicit a lipogenic response greater than the solvent controls.
[00266] Based upon their adipogenic potential in 3T3-L1 cells, the positive hops phytochemical genera in this study, which included isomerized alpha acids, alpha acids and beta acids as well as xanthohumols, may be expected to increase insulin sensitivity and decrease serum triglycerides in humans or other animals exhibiting signs or symptoms of insensitivity to insulin.
Example 21
Hops phytochemicals increase adiponectin secretion in insulin-resistant 3T3-L1 adipocytes.
[00267] The Model — The 3T3-L1 murine fibroblast model as described in Examples
11 and 12 were used in this example. Standard chemicals, hops compounds RIAA, IAA, THIAA, HHIAA, xanthohumols, hexahydrocolupulone, spent hops were as described, respectively, in Examples 12 and 20.
[00268] Cell Culture and Treatment — Cells were cultured as described in Example 12 and treated with hops phytochemicals as previously described. Adiponectin assays and statistical interpretations were as described in Example 12. Potency of the test materials was estimated using a modification of the method of Hofstee for determination of the apparent Michaelis constants and maximum velocities. Substituting {relative adiponectin secretion/[concentration]} for the independent variable v/[S] and {relative adiponectin secretion} for the dependant variable {v}, produced a relationship of the form y = mx + b. Maximum adiponectin secretion relative to the solvent control was estimated from the y- intercept, while the concentration of test material necessary for half maximal adiponectin secretion was computed from the negative value of the slope.
[00269] Results - The positive control troglitazone maximally enhanced adiponectin secretion 2.44-fold at 2.5 μg/ml over the solvent control in insulin-resistant 3T3-L1 cells (Figure 19). All hops phytochemicals tested demonstrated enhanced adiponectin secretion relative to the solvent control, with isoalpha acids producing significantly more adiponectin secretion than troglitazone (2.97-fold relative to controls). Of the four doses tested, maximal adiponectin secretion was observed at 5 μg/ml, the highest dose, for isoalpha acids, Rho isoalpha acids, hexahydroisoalpha acids and tetrahydroisoalpha acids. For xanthohumols, spent hops and hexahydro colupulone the maximum observed increase in adiponectin secretion was seen at 1.25, 25 and 12.5 μg/ml, respectively. Observed maximal relative adiponectin expression was comparable to troglitazone for xanthohumols, Rho isoalpha acids, and spent hops and less than troglitazone, but greater than control, for hexahydroisoalpha acids, hexahydro colupulone and tetrahydroisoalpha acids.
Table 20
Maximum adiponectin secretion and concentration of test material necessary for half maximal adiponectin secretion estimated, respectively, from the v-intercept and slope of
Hofstee plots.
Maximum Adiponectin Secretion/1' Test Material at Half Maximal Secretion Test Material [Fold relative to control] [μg/mL]
Isoalpha acids 3.17 0.49
Xanthohumol 2.47 0.037
Rho isoalpha acids 2.38 0.10
Troglitazone121 2.29 0.0B5
Spent hops 2.21 2.8
Hexahydroisoalpha acidspl 1.89 0.092
Hexahydro colupulone121 1.83 3.2
Tetrahydroisoalpha acids 1.60 0.11
[1]Estimated from linear regression analysis of Hofstee plots using all four concentrations tested [2]One outlier omitted and three concentrations used for dose-response estimates
[00270] As seen in Table 20, estimates of maximal adiponectin secretion derived from modified Hofstee plots (Figure 20) supported the observations noted above. y-Intercept estimates of maximum adiponectin secretion segregated roughly into three groups: (1) isoalpha acids, (2) xanthohumols, Rho isoalpha acids, troglitazone, and spent hops, and (3) hexahydroisoalpha acids, hexahydro colupulone and tetrahydroisoalpha acids. The concentration of test material required for stimulation of half maximal adiponectin secretion in insulin-resistant 3T3-L1 cells, approximately 0.1 μg/ml, was similar for troglitazone, Rho isoalpha acids, tetrahydroisoalpha acid and hexahydroisoalpha acids. The concentration of isoalpha acids at half maximal adiponectin secretion 0.49 μg/ml was nearly 5-fold greater. Xanthohumols exhibited the lowest dose for half maximal adiponectin secretion estimated at 0.037 μg/ml. The highest concentrations for the estimated half maximal adiponectin secretion variable were seen for spent hops and hexahydro colupulone, respectively, 2.8 and 3.2 μg/ml. [00271] Based upon their ability to enhance adiponectin secretion in insulin-resistant
3T3-L1 cells, the positive hops phytochemical genera seen in this study, isoalpha acids, Rho- isoalpha acids, tetrahydroisoalpha acids, hexahydroisoalpha acids, xanthohumols, spent hops and hexahydro colupulone, may be expected to have a positive effect on all clinical pathologies in which plasma adiponectin concentrations are depressed.
Example 22
Hops phytochemicals exhibit anti-inflammatory activity through enhanced adiponectin secretion and inhibition of interleukin-6 secretion in insulin-resistant 3T3-L1 adipocytes.
(00272] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Adiponectin and IL-6 were assayed as described, respectively in Examples 12 and 18. Standard chemicals, hops compounds RIAA, IAA, THIAA, HHIAA, xanthohumols, hexahydrocolupulone, spent hops were as described in Examples 12 and 20.
[00273] Statistical Calculations and Interpretation — All assays were preformed in duplicate. For statistical analysis, the effect of hops derivatives on adiponectin or IL-6 secretion was computed relative to the solvent control. Differences among the doses were determined using analysis of variance without correction for multiple comparisons; the nominal five percent probability of a type I error was selected.
[00274] Results - Troglitazone and all hops derivatives tested increased adiponectin secretion in the presence of high concentrations of insulin (Table 21). Troglitazone did not decrease IL-6 secretion in this model. In fact, troglitazone, and HHCL exhibited two concentrations in which IL-6 secretion was increased, while THIAA and spent hops increased IL-6 at the highest concentration and had no effect at the other concentrations. The effect of other hops derivatives on IL-6 secretion was generally biphasic. At the highest concentrations tested, RIAA, HHIAA, and XN increased IL-6 secretion; only IAA did not. Significant decreases in IL-6 secretion were noted for RIAA, IAA, THIAA, and XN.
Table 21
Effect of hops compounds on adiponectin and interleukin-6 secretion insulin-resistant 3T3-L1 adipocytes.
Concentration |
Figure imgf000087_0001
The Acacia catechu test material or iπdomethaciπ was added in concert with 166 nM insulin to D5 3T3-L1 adipocytes. On the following day, supernatant media were sampled for adiponectin, IL-6 and resistin determination. All values were indexed to the insulin only control. t Adiponectin Index = [Adiponectin]τcst/[Adiponectin]insuijn control ffIL-6 Index = [IL-6Test]/[IL-6insuiin control]
*Index value is mean ± 95% confidence interval computed from residual mean square of the analysis of variance. For adiponectin or adiponectin/IL-6, values < 0.7 or > 1.3 are significantly different from insulin control and for IL-6, values <0.77 or >1.23 are significantly different from insulin control.
#Significantly different from insulin control p<0.05.
[00275] The adiponectin/IL-6 ratio, a metric of overall anti-inflammatory effectiveness, was strongly positive (>2.00) for RIAA, IAA HHIA, and XN. THIAA, HHCL and spent hops exhibited positive, albeit lower, adiponectin/IL-6 ratios. For troglitazone the adiponectin/IL-6 ratio was mixed with a strongly positive response at 2.5 and 0.625 μg/ml and no effect at 5.0 or 1.25 μg/ml.
[00276] The data suggest that the pro-inflammatory effect of hyperinsulinemia can be attenuated in adipocytes by hops derivatives RIAA, IAA, HHIA, THIAA, XN, HHCL and spent hops. In general, the anti-inflammatory effects of hops derivatives in hyperinsulinemia conditions hyperinsulinemia uncomplicated by TNFa were more consistent than those of troglitazone.
Example 23
Hops phytochemicals increase adiponectin secretion in TNFotreated 3T3-L1 adipocytes.
[00277] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals and hops compounds IAA, RIAA, HHIAA, and THIAA, were as described, respectively, in Examples 13 and 20. Hops derivatives were tested at concentrations of 0.625, 1.25, 2.5, and 5.0 μg/ml. Adiponectin was assayed as described in Example 12.
[00278] Results - Overnight treatment of day 5 (D5) 3T3-L1 adipocytes with 10 ng
TNFoVmI markedly suppressed adiponectin secretion (Figure 21). The hops derivatives IAA, RIAA, HHIAA and THIAA all increased adiponectin secretion relative to the TNFoysolvent control. Linear dose-response curves were observed with RIAA and HHIAA resulting in maximal inhibition at the highest concentration tested 5.0 μg/ml. IAA elicited maximal secretion of adiponectin at 1.25 μg/ml, while THIAA exhibited a curvilinear response with maximal adiponectin secretion at 5.0 μg/ml.
[00279] The ability of hops derivatives IAA, RIAA, HHIAA and THIAA to increase adipocyte adiponectin secretion in the presence of supraphysiological concentrations of TNFα supports the usefulness of these compounds in the prevention or treatment of inflammatory conditions involving suboptimal adipocyte functioning.
Example 24
Acacia catechu formulation synergistic interaction with hops derivatives to alter lipogenesis and adiponectin secretion in 3T3-L1 adipocytes.
[00280] The Model - The 3T3-L1 murine fibroblast model as described in Examples 11 and 13 was used in these experiments.
[00281] Test Chemicals and Treatment - Standard chemicals used were as noted in
Examples 11 and 13. 3T3-L1 adipocytes were treated prior to differentiation as in Example 11 for computing the lipogenic index or with TNFα as described in Example 12 for assessing the adiponectin index. Acacia catechu sample #5669 as described in Example 14 was used with hops derivatives Rho-isoalpha acids and isoalpha acids as previously described. Acacia catechu and the 5:1 and 10:1 combinations of Acacia:RIAA and Acacia:IAA were tested at 50, 10, 5.0 and 1.0 μg/ml. RIAA and IAA were tested independently at 5.0, 2.5, 1.25 and 0.625 μg/ml.
[00282] Calculations — Estimates of expected lipogenic response and adiponectin secretion of the Acacia/hops combinations and determination of synergy were made as previously described.
[00283] Results - All combinations tested exhibited lipogenic synergy at one or more concentrations tested (Table 22). Acacia:RIAA combinations were generally more active than the Acacia:IAA combinations with Acacia:RIAA [5:1] demonstrating synergy at all doses and Acacia:RIAA [10:1] synergistic at 10 and 5.0 μg/ml and not antagonistic at any concentration tested. The Acacia:IAA [10:1] combination was also synergistic at the two mid-doses and showed no antagonism. While Acacia:IAA [5:1] was synergistic at the 50 μg/ml concentration, it was antagonistic at the 5.0 μg/ml dose.
[00284] Similarly, all combinations demonstrated synergy with respect to increasing adiponectin secretion at one or more concentrations tested (Table 23). Acacia:IAA [10:1] exhibited synergy at all doses, while Acaca:RIAA [5:1] and Acacia:RIAA [10:1] were synergistic at three doses and antagonistic at one concentration. The AcaciarIAA [5:1] combination was synergistic at 1.0 μg/ml and antagonistic at the higher 10 μg/ml.
Table 22
Observed and expected lipogenic response elicited by Acacia catechu and hops derivatives in the insulin-resistant 3T3-1 model.
Figure imgf000090_0001
fLipogenic Index = [OD]TeSt/[OD]DMso control- l)Upper 95% confidence limit is 1.03 with least significant difference = 0.03. 2)Upper 95% confidence limit is 1.03 with least significant difference = 0.03 3)Upper 95% confidence limit is 1.07 with least significant difference = 0.07. 4)Upper 95% confidence limit is 1.02 with least significant difference = 0.02.
Table 23
Observed and expected adiponectin secretion elicited by Acacia catechu and hops derivatives in the TNFα/3T3-l model.
Figure imgf000090_0002
Figure imgf000091_0001
fAdiponectin Index = [Adiponectin]τest/[Adiponectin]τNFα control l)Upper 95% confidence limit is 1.07 with least significant difference = 0.07.
2)Upper 95% confidence limit is 1.03 with least significant difference = 0.03
[00285] Combinations of Acacia catechu and the hops derivatives Rho isoalpha acids or isoalpha acids exhibit synergistic combinations and only few antagonistic combinations with respect to increasing lipid incorporation in adipocytes and increasing adiponectin secretion from adipocytes.
Example 25
Anti-inflammatory activity of hops derivatives in the lipopolysaccharide/3T3-Ll adipocyte model.
[00286] The Model — The 3T3-L1 murine adipocyte model as described in Examples
11 and 13 was used in these experiments.
[00287] Test Chemicals and Treatment - Standard chemicals were as noted in
Examples 11 and 13, however, 100 ng/ml of bacterial lipopolysaccharide (LPS, Sigma, St. Louis, MO) was used in place of TNFα on D5. Hops derivatives Rho-isoalpha acids and isoalpha acids used were as described in Example 20. The non-steroidal anti-inflammatory drugs (NSAIDs) aspirin, salicylic acid, and ibuprofen were obtained from Sigma. The commercial capsule formulation of celecoxib (Celebrex™, G.D. Searle & Co. Chicago, IL) was used and cells were dosed based upon content of active ingredient. Hops derivatives, ibuprofen, and celecoxib were dosed at 5.00, 2.50, 1.25 and 0.625 μg/ml. Indomethacin, troglitazone, and pioglitazone were tested at 10, 5.0, 1.0 and 0.50 μg/ml. Concentrations for aspirin were 100, 50.0, 25.0 and 12.5 μg/ml, while those for salicylic acid were 200, 100, 50.0 and 25.0 μg/ml. IL-6 and adiponectin were assayed and data were analyzed and tabulated as previously described in Example 18 for IL-6 and Example 13 for adiponectin.
[00288] Results - LPS provided a 12-fold stimulation of IL-6 in D5 adipocytes. All test agents reduced IL-6 secretion by LPS-stimulated adipocytes to varying degrees. Maximum inhibition of IL-6 and concentrations for which this maximum inhibition were observed are presented in Table 24. Due to a relatively large within treatment variance, the extent of maximum inhibition of IL-6 did not differ among the test materials. The doses for which maximum inhibition occurred, however, did differ considerably. The rank order of potency for IL-6 inhibition was ibuprofen > RIAA = IAA > celecoxib > pioglitazone = indomethacin > troglitazone > aspirin > salicylic acid. On a qualitative basis, indomethacin, troglitazone, pioglitazone, ibuprofen and celecoxib inhibited IL-6 secretion at all concentrations tested, while RIAA, IAA, and aspirin did not significantly inhibit IL-6 at the lowest concentrations (data not shown).
[00289] LPS treatment of D5 3T3-L1 adipocytes decreased adiponectin secretion relative to the DMSO control (Table 25). Unlike IL-6 inhibition in which all test compounds inhibited secretion to some extent, aspirin, salicylic acid and celecoxib failed to induce adiponectin secretion in LPS-treated 3T3-L1 adipocytes at any of the does tested. Maximum adiponectin stimulation of 15, 17, 20 and 22% was observed, respectively, for troglitazone, RIAA, IAA and ibuprofen at 0.625 μg/ml. Pioglitazone was next in order of potency with adiponectin stimulation of 12% at 1.25 μg/ml. With a 9% stimulation of adiponectin secretion at 2.50 μg/ml, indomethacin was least potent of the active test materials.
[00290] In the LPS/3T3-L1 model, hops derivatives RIAA and IAA as well as ibuprofen decreased IL-6 secretion and increased adiponectin secretion at concentrations likely to be obtained in vivo. The thiazolidinediones troglitazone and pioglitazone were less potent as inhibitors of IL-6 secretion, requiring higher doses than hops derivatives, but similar to hops derivatives with respect to adiponectin stimulation. No consistent relationship between anti-inflammatory activity in macrophage models and the adipocyte model was observed for the NSAIDs indomethacin, aspirin, ibuprofen and celecoxib.
Table 24 Maximum inhibition of IL-6 secretion in LPS/3T3-L1 adipocytes by hops derivatives and selected NSAJDs
Figure imgf000093_0001
The test materials were added in concert with 100 ng LPS/ml to D5 3T3-L1 adipocytes. On the following day, supernatant media were sampled for JL-6 determination. All values were indexed to the LPS control as noted below. Concentrations presented represent dose providing the maximum inhibition of IL-6 secretion and those values less than 0.70 are significantly (p<0.05) less than the LPS control. fIL-6 Index = [IL-6Test - IL-6ntroi]/[IL-6Lps - IL-6ntroi] *Significantly different from LPS control p<0.05).
Table 25
Maximum stimulation of adiponectin secretion in LPS/3T3-L1 adipocytes by hops derivatives and selected NSAIDs
Figure imgf000093_0002
f Adiponectin Index = [Adiponectin]τest/[Adiponectin]Lps control
* Values greater than 1.07 are significantly different from LPS control p<0.05). NS = not significantly different from the LPS control.
Example 26
Synergy of Acacia catechu or hops derivatives in combination with curcumin or xanthohumols in the TNFo/3T3-l model.
[00291] The Model — The 3T3-L1 murine fibroblast model as described in Examples
11 and 13 was used in these experiments.
[00292] Test Chemicals and Treatment - Standard chemicals used were as noted in
Example 11 and 13. 3T3-L1 adipocytes were stimulated with TNFo; as described in Example 13 for assessing the adiponectin index. Acacia catechu sample #5669 as described in Example 14, hops derivatives Rho-isoalpha acids and xanthohumol as described in Example 20, and curcumin as provided by Metagenics (Gig Harbor, WA) and were used in these experiments. Acacia catechu and the 5:1 combinations of
Figure imgf000094_0001
and ^cαc/α:xanthohumol were tested at 50, 10, 5.0 and 1.0 μg/ml. RIAA and the 1:1 combinations with curcumin and XN were tested at 10, 5, 1.0 and 0.50 μg/ml.
[00293] Calculations — Estimates of expected adiponectin index of the combinations and determination of synergy were made as described previously.
[00294] Results — TNFα reduced adiponectin secretion to about 50 percent of solvent only controls. The positive control pioglitazone increased adiponectin secretion by 80 percent (Table 26). Combinations of Acacia with curcumin or XN proved to be antagonistic at the higher concentrations and synergistic at the lower concentrations. Similarly, RIAA and curcumin were antagonistic at the three higher doses, but highly synergistic at the lowest dose 1.0 μg/ml. The two hops derivative RIAA and XN did not demonstrate synergy in adiponectin secretion from TNFor-stimulated 3T3-L1 cells.
[00295] In TNFα-treated 3T3-L1 adipocytes, both Acacia and RIAA synergistically increased adiponectin secretion, while only Acacia demonstrated synergy with XN.
Table 26
Synergy of Acacia catechu and hops derivatives in combinations with curcumin or xanthohumols in the TNFα/3T3-l model. W
Figure imgf000095_0001
f Adiponectin Index = [ Adiponectin]τcs/[ Adi ponectin]τNF<* control
1) 95% confidence limits are 0.961 to 1.049 with least significant difference = 0.049.
Example 27
In vitro synergy of lipogenesis by conjugated linoleic acid in combination with hops derivative Rho-isoalpha acids in the insulin-resistant 3T3-L1 adipocyte model.
[00296J The Model - The 3T3-L1 murine fibroblast model as described in Examples 11 and 13 was used in these experiments.
[00297] Test Chemicals and Treatment - Standard chemicals used were as noted in
Example 11. 3T3-L1 adipocytes were treated prior to differentiation as in Example 11 for computing the lipogenic index. Powdered CLA was obtained from Lipid Nutrition (Channahon, IL) and was described as a 1:1 mixture of the c9tl 1 and 110c 12 isomers. CLA and the 5:1 combinations of CLArRIAA were tested at 50, 10, 5.0 and 1.0 μg/ml. RIAA was tested at 10, 1.0 and 0.1 μg/ml for calculation of expected lipogenic index as described previously. [00298] Results - RIAA synergistically increased triglyceride content in combination with CLA. Synergy was noted at all does (Table 27).
[00299] Synergy between CLA and RIAA was observed over a wide range of doses and potentially could be used to increase the insulin sensitizing potency of CLA.
Table 27
Synergy of lipogenesis by conjugated linoleic acid in combination Rho-isoalpha acids in the insulin-resistant 3T3-L1 adipocyte model.
Figure imgf000096_0001
tLipogenic Index = [OD]Test/[OD]DMso control-
1) Upper 95% confidence limit is 1.05 with least significant difference = 0.05.
Example 28
Hops phvtochemicals inhibit NF-kB activation in TNFα-treated 3T3-L1 adipocytes.
[00300] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments.
[00301] Cell Culture and Treatment - Following differentiation 3T3-L1 adipocytes were maintained in post-differentiation medium for an additional 40 days. Standard chemicals, media and hops compounds RIAA and xanthohumol were as described in Examples 13 and 20. Hops derivatives and the positive control pioglitazone were tested at concentrations of 2.5, and 5.0 μg/ml. Test materials were added 1 hour prior to and nuclear extracts were prepared three and 24 hours following treatment with TNFα.
[00302] ELISA - 3T3-L1 adipocytes were maintained in growth media for 40 days following differentiation. Nuclear NF-kBp65 was determined using the TransAM™ NF-kB kit from Active Motif (Carlsbad, CA) was used with no modifications. Jurkat nuclear extracts provided in the kit were derived from cells cultured in medium supplemented with 50 ng/ml TPA (phorbol, 12-myri state, 13 acetate) and 0.5 μM calcium ionophore A23187 for two hours at 37°C immediately prior to harvesting.
[00303] Protein assay — Nuclear protein was quantified using the Active Motif
Fluorescent Protein Quantification Kit.
[00304] Statistical Analysis — Comparisons were performed using a one-tailed
Student's t-test. The probability of a type I error was set at the nominal five percent level.
[00305] Results — The TPA-treated Jurkat nuclear extract exhibited the expected increase in NF-kBp65 indicating adequate performance of kit reagents (Figure 22). Treatment of D40 3T3-L1 adipocytes with 10 ng TNFα/ml for three (Figure 22A) or 24 hours (Figure 22B), respectively, increased nuclear NF-kBp65 2.1- and 2.2-fold. As expected, the PPAR.7 agonist pioglitazone did not inhibit the amount of nuclear NF-kBp65 at either three or 24 hours following TNFα treatment. Nuclear translocation of NF-kBp65 was inhibited, respectively, 9.4 and 25% at 5.0 and 2.5 μg RIAA/ml at three hours post TNFα. At 24 hours, only the 5.0 RIAA/ml treatment exhibited significant (p<0.05) inhibition of NF-kBp65 nuclear translocation. Xanthohumols inhibited nuclear translocation of NF-kBp65, respectively, 15.6 and 6.9% at 5.0 and 2.5 μg/ml at three hours post-TNFo: treatment and 13.4 and 8.0% at 24 hours.
[00306] Both RIAA and xanthohumols demonstrated consistent, albeit small, inhibition of nuclear translocation of NF-kBp65 in mature, insulin-resistant adipocytes treated with TNFa. This result differs from that described for PP ARγ agonists, which have not been shown to inhibit nuclear translocation of NF-kBp65 in 3T3-L1 adipocytes.
Example 29
Acacia catechu extract and metformin synergistically increase triglyceride incorporation in insulin resistant 3T3-L1 adipocytes.
[00307] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. All chemicals and procedures used were as described in Example 11.
[00308] Test Chemicals and Treatment - Metformin was obtained from Sigma (St.
Louis, MO). Test materials were added in dimethyl sulfoxide at Day 0 of differentiation and every two days throughout the maturation phase (Day 6/7). As a positive control, troglitazone was added to achieve a final concentration of 4.4 μg/ml. Metformin, Acacia catechu sample #5669 and the metfoπnin/Acacia combination of 1:1 (w/w) were tested at 50 μg test material/ml. Differentiated 3T3-L1 cells were stained with 0.2% Oil Red O. The resulting stained oil droplets were dissolved with isopropanol and quantified by spectrophotometric analysis at 530 nm. Results were represented as a relative triglyceride content of fully differentiated cells in the solvent controls.
[00309] Calculations - An estimate of the expected adipogenic effect of the metformin/
Acacia catechu extract was made using the relationship: 1/LI = X/LIx + Y/LIy, where LI = the lipogenic index, X and Y were the relative fractions of each component in the test mixture and X + Y = 1. Synergy was inferred if the mean of the estimated LI fell outside of the 95% confidence interval of the estimate of the corresponding observed fraction. This definition of synergy, involving comparison of the effects of a combination with that of each of its components, was described by Berenbaum [Berenbaum, M. C. What is synergy? Pharmacol Rev 41(2), 93-141, (1989)].
[00310] Results - The Acacia catechu extract was highly lipogenic, increasing triglyceride content of the 3T3-L1 cells by 32 percent (Figure 23) yielding a lipogenic index of 1.32. With a lipogenic index of 0.79, metformin alone was not lipogenic. The metformin/4 cacia catechu extract combination demonstrated an observed lipogenic index of 1.35. With an expected lipogenic index of 98, the metformin/ Acacia catechu extract demonstrated synergy as the observed lipogenic index fell outside of the two percent 95% upper confidence limit for the expected value.
[00311] Based upon the lipogenic potential demonstrated in 3T3-L1 cells, 1 :1 combinations of metformin and Acacia catechu extract would be expected to behave synergistically in clinical use. Such combinations would be useful to increase the range of positive benefits of metformin therapy such as decreasing plasma triglycerides or extending the period of metformin efficacy.
Example 30
In vitro synergies of lipogenesis by hops derivatives and thiazolidinediones in the insulin- resistant 3T3-L1 adipocyte model. [00312] The Model - The 3T3-L1 murine fibroblast model as described in Examples
11 and 13 was used in these experiments.
[00313] Test Chemicals and Treatment - Standard chemicals used were as noted in
Example 11. 3T3-L1 adipocytes were treated prior to differentiation as in Example 11 for computing the lipogenic index. Troglitazone was obtained from Cayman Chemicals (Chicago, IL). Pioglitazone was obtained as the commercial, tableted formulation (ACTOSE®, Takeda Pharmaceuticals, Lincolnshire, IL). The tablets were crushed and the whole powder was used in the assay. All results were computed based upon active ingredient content. Hops derivatives Rho-isoalpha acids and isoalpha acids used were as described in Example 20. Troglitazone in combination with RIAA and IAA was tested at 4.0 μg/ml, while the more potent pioglitazone was tested in 1 :1 combinations with RIAA and IAA at 2.5 μg/ml. All materials were also tested independently at 4.0 and 2.5 μg/ml for calculation of expected lipogenic index as described in Example 34.
[00314] Results — When tested at 4.0 and 2.5 μg/ml, respectively, with troglitazone or piroglitazone, both Rho-isoalpha acids and isoalpha acids increased triglyceride synthesis synergistically with the thiazolidinediones in the insulin-resistant 3T3-L1 adipocyte model (Table 28).
[00315] Hops derivatives Rho-isoalpha acids and isoalpha acids could synergistically increase the insulin sensitizing effects of thiazolidinediones resulting in potential clinical benefits of dose-reduction or increased numbers of patients responding favorably.
Table 28
In vitro synergies of hops derivatives and thiazolidinediones in the insulin-resistant 3T3-L1 adipocyte model.
Figure imgf000099_0001
fLipogenic Index = [OD]Test/[OD]DMso control-
1) Upper 95% confidence limit is 1.02 with least significant difference = 0.02.
2) Upper 95% confidence limit is 1.05 with least significant difference = 0.05. Example 31
In vitro synergies of Rho-isoalpha acids and metformin in the TNFα^3T3-Ll adipocyte model.
[00316] The Model - The 3T3-L1 murine fibroblast model as described in Example 11 was used in these experiments. Standard chemicals used and treatment of adipocytes with 10 ng TNI5WmI were as noted, respectively, in Examples 11 and 13.
[00317] Test Materials and Cell Treatment — Metformin was obtained from Sigma (St.
Louis, MO) and Rho-isoalpha acids were as described in Example 20. Metformin at 50, 10, 5.0 or 1.0 μg/ml without or with 1 μg RIAA/ml was added in concert with 10 ng TNFo/ml to D5 3T3-L1 adipocytes. Culture supernatant media were assayed for IL-6 on Day 6 as detailed in Example 11. An estimate of the expected effect of the metformin :RIAA mixtures on IL-6 inhibition was made as previously described.
[00318] Results - TNFa provided a six-fold increase in IL-6 secretion in D5 adipocytes. Troglitazone at 1 μg/ml inhibited IL-6 secretion 34 percent relative to the controls, while 1 μg RIAA inhibited DL-6 secretion 24 percent relative to the controls (Table 29). Metformin in combination with 1 μg RIAA/ml demonstrated synergy at the 50 μg/ml concentration and strong synergy at the 1 μg/ml concentration. At 50 μg metformin/ml, 1 μg RIAA provided an additional 10 percent inhibition in the mixture; while at 1 μg metformin, 1 μg RIAA increased IL-6 inhibition by 35 percent. Antagonism and no effect, respectively, were seen of the metformin:RIAA combinations at the two mid-doses.
[00319] Combinations of metformin and Rho-isoalpha acids function synergistically at both high and low concentrations to reduce IL-6 secretion from TNFor-treated 3T3-L1 adipocytes.
Table 29
Synergistic inhibition of IL-6 secretion in TNFo/3T3-Ll adipocytes by hops Rho-isoalpha acids and metformin.
Figure imgf000100_0001
Figure imgf000101_0001
The test materials were added in concert with 10 ng TNTWmI to D5 3T3-L1 adipocytes at the stated concentrations. On the following day, supernatant media were sampled for IL-6 determination. All values were indexed to the TNFα control. fIL-6 Index = [IL-6Test - IL-6Controi]/[IL-6τNFα - IL-6controi]
* Values less than 0.93 are significantly (p<0.05) less than the TNFa control.
Example 32
Effects of test compounds on cancer cell proliferation in vitro
[00320] This experiment demonstrates the direct inhibitory effects on cancer cell proliferation in vitro for a number of test compounds of the instant invention.
[0032 IJ Methods - The inhibitory effects of test compounds of the present invention on cancer cell proliferation were examined in the RL 95-2 endometrial cancer cell model (an over expressor of AKT kinase), and in the HT-29 (constitutively expressing COX-2) and SW480 (constitutively expressing activated AKT kinase) colon cancer cell models. Briefly, the target cells were plated into 96 well tissue culture plates and allowed to grow until subconfluent. The cells were then treated for 72 hours with various amounts of the test compounds as described in Example 4 and relative cell proliferation determined by the CyQuant (Invitrogen, Carlsbad, CA) commercial fluorescence assay.
[00322] Results - RL 95-2 cells were treated for 72 hours with 10 μg/ml of MgDHIAA
(mgRho), IAA, THIAA, TH-HHIAA ( a 1 :1 mixture of THIAA & HHIAA), Xn (xanthohumol), LY (LY 249002, a PI3K inhibitor), EtOH (ethanol), alpha (alpha acid mixture), and beta (beta acid mixture). The relative inhibition on cell proliferation is presented as Figure 24, showing a greater than 50% inhibition for xanthohumol relative to the DMSO solvent control. Figures 25 & 26 display the dose response results for various concentrations of RIAA or THIAA on HT-29 and SW480 cancer cells respectively. Median inhibitory concentrations for RIAA and THIAA were 31 and 10 μM for the HT-29 cell line and 38 and 3.2 μM for the SW480 cell line.
Example 33
In vivo hypoglycemic action of Acacia nilotica and hops derivatives in the KK-Ay Mouse diabetes model.
[00323] The Model - Male, nine-week old KK-Ay/Ta mice averaging 40 ± 5 grams were used to assess the potential of the test materials to reduce fasting serum glucose or insulin concentrations. This mouse strain is the result of hybridization between the KK strain, developed in the 1940s as a model of diabetes and a strain of Ay/a genotype. The observed phenotype is the result of polygenic mutations that have yet to be fully characterized but at least four quantitative trait loci have been identified. One of these is linked to a missense mutation in the leptin receptor. Despite this mutation the receptor remains functional although it may not be fully efficient. The KK strain develops diabetes associated with insensitivity to insulin and glucose intolerance but not overt hyperglycemia. Introduction of the Ay mutation induces obesity and hyperglycemia. The Ay mutation is a 170kb deletion of the RaIy gene that is located 5' to the agouti locus and places the control for agouti under the RaIy promoter. Homozygote animals die before implantation.
(00324] Test Materials — Acacia nilotica sample #5659 as described in Example 14 and hops derivatives Rho-isoalpha acids, isoalpha acids and xanthohumols as described in Example 20 were used. The Acacia nilotica, RIAA and IAA were administered at 100 mg/kg/day, while XN was dosed at 20 mg/kg. Additionally, 5:1 and 10:1 combinations of Acacia nilotica with RIAA, IAA and XN were formulated and dosed at 100 mg/kg/day.
[00325] Testing Procedure - Test substances were administered daily by gavage in
0.2% Tween-80 to five animals per group. Serum was collected from the retroorbital sinus before the initial dose and ninety minutes after the third and final dose. Non-fasting serum glucose was determined enzymatically by the mutarotase/glucose oxidase method and serum insulin was determined by a mouse specific ELISA (enzyme linked immunosorbent assay).
[00326] Data Analysis — To assess whether the test substances decreased either serum glucose or insulin relative to the controls, the post-dosing glucose and insulin values were first normalized relative to pre-dosing concentrations as percent pretreatment for each mouse. The critical value (one-tail, lower 95% confidence interval for the control mice) for percent pretreatment was computed for both the glucose and insulin variables. Each percent pretreatment value for the test materials was compared with the critical value of the control. Those percent pretreatment values for the test materials that were less than the critical value for the control were considered significantly different (p<0.05) from the control.
[00327] Results — During the three-day treatment period, non-fasting, serum glucose rose 2.6% while serum insulin decreased 6.7% in control mice. Rosigltiazone, Acacia nilotica, XN:Acacia [1:5], XNrAcacia [1:10], Acacia:RIAA [5:1], xanthohumols, Acacia:IAA [5:1], isomerized alpha acids and Rho-isoalpha acids all decreased non-fasting serum glucose relative to the controls with no effect on serum insulin. Acacia:RIAA [10:1] and Acacia:IAA [10:1] had no effect on either serum glucose or insulin (Table 30).
[00328] The rapid hypoglycemic effect of Acacia nilotica sample #5659, xanthohumols, isomerized alpha acids, Rho-isoalpha acids and their various combinations in the KK-Ay mouse model of type 2 diabetes supports their potential for clinical efficacy in the treatment of human diseases associated with hyperglycemia.
Table 30
Effect of Acacia nilotica and hops derivatives on non-fasting serum glucose and insulin in
KK-Ay diabetic mice.
Figure imgf000103_0001
fDosing was performed once daily for three consecutive days on five animals per group. #Signifϊcantly less than control (p<0.05). Example 34
In vivo synergy of Acacia nilotica and hops derivatives in the diabetic db/db mouse model.
[00329J The Model - Male, C57BLKS/J m+/m+ Lepr* (db/db) mice were used to assess the potential of the test materials to reduce fasting serum glucose or insulin concentrations. This strain of mice is resistant to leptin by virtue of the absence of a functioning leptin receptor. Elevations of plasma insulin begin at 10 to 14 days and of blood sugar at 4 to 8 weeks. At the time of testing (9 weeks) the animals were markedly obese 50 ± 5 g and exhibited evidence of islet hypertrophy.
[00330] Test Materials — The positive controls metformin and rosiglitazone were dosed, respectively, at 300 mg/kg-day and 1.0 mg/kg-day for each of three consecutive days. Acacia nilotica sample #5659, hops derivatives and their combinations were dosed as described previously.
[00331] Testing Procedure - Test substances were administered daily by gavage in
0.2% Tween-80. Serum was collected from the retroorbital sinus before the initial dose and ninety minutes after the third and final dose. Non-fasting serum glucose was determined eπzymatically by the mutarotase/glucose oxidase method and serum insulin was determined by a mouse specific ELISA.
[00332] Results — The positive controls metformin and rosiglitazone decreased both serum glucose and insulin concentrations relative to the controls (Table 31). Only RIAA and XN demonstrated acceptable results as single test materials. RIAA reduced serum insulin, while XN produced a reduction in serum glucose with no effect on insulin. Acacia:RIAA [5:1] was the most effective agent tested for reducing serum insulin concentrations, providing a 21 percent reduction in serum insulin levels versus a 17 percent reduction in insulin concentrations by the biguanide metformin and a 15 percent decrease by the thiazolidinedione rosiglitazone. The response of this Acacia:RIAA [5:1] combination was greater than the responses of either individual component thus exhibiting a potential for synergy. Acacia nilotica alone failed to reduce either serum glucose or insulin, while RIAA reduced serum insulin to a similar extent as metformin. Of the remaining test materials, the Acacia:IAA [10:1] combination was also effective in reducing serum insulin concentrations. [00333] The rapid reduction of serum insulin affected by Rho-isoalpha acids and reduction of serum glucose by xanthohumols in the db/db mouse model of type 2 diabetes supports their potential for clinical efficacy in the treatment of human diseases associated with insulin insensitivity and hyperglycemia. Further, the 5:1 combination of Rho-isoalpha acids and Acacia catechu appeared synergistic in the db/db murine diabetes model. The positive responses exhibited by Rho-isoalpha acids, xanthohumols and the AcaciarRIAA [5:1] formulation in two independent animal models of diabetes and three in vitro models supports their potential usefulness in clinical situations requiring a reduction in serum glucose or enhance insulin sensitivity.
Table 31
Effect of Acacia nilotica and hops derivatives on non-fasting serum glucose and insulin in db/db diabetic mice.
Figure imgf000105_0001
f Dosing was performed once daily for three consecutive days on five animals per group. #Significantly less than respective control (p<0.05).
Example 35
In vivo optimization of Acacia nilotica and hops derivative ratio in the diabetic db/db mouse model.
[00334] The Model - Male, C57BLKS/J m+/m+ heprdb (db/db) mice were used to assess the potential of the test materials to reduce fasting serum glucose or insulin concentrations. This strain of mice is resistant to leptin by virtue of the absence of a functioning leptin receptor. Elevations of plasma insulin begin at 10 to 14 days and of blood sugar at 4 to 8 weeks. At the time of testing (9 weeks) the animals were markedly obese 50 ± 5 g and exhibited evidence of islet hypertrophy.
[00335J Test Materials - The positive controls metformin and rosiglitazone were dosed, respectively, at 300 mg/kg-day and 1.0 mg/kg-day for each of five consecutive days. The hops derivative RIAA and Acacia nilotica sample #5659 in ratios of 1 :99, 1 :5, 1:2, 1:1, 2:1, and 5:1 were dosed at 100 mg/kg.
[00336] Testing Procedure - Test substances were administered daily by gavage in
0.2% Tween-80. Serum was collected from the retroorbital sinus before the initial dose and ninety minutes after the fifth and final dose. Non-fasting serum glucose was determined enzymatically by the mutarotase/glucose oxidase method and serum insulin was determined by a mouse specific ELISA.
[00337] Results - The positive controls metformin and rosiglitazone decreased both serum glucose and insulin concentrations relative to the controls (p<0.05, results not shown). Individually, RIAA and Acacia at 100 mg/kg for five days reduced serum glucose, respectively, 7.4 and 7.6 percent relative to controls (p<0.05). Combinations of RIAA and Acacia at 1 :99, 1 :5 or 1 :1 appeared antagonistic, while 2:1 and 5:1 ratios of RIAA:Acacia decreased serum glucose, respectively 11 and 22 percent relative to controls. This response was greater than either RIAA or Acacia alone and implies a synergic effect between the two components. A similar effect was seen with decreases in serum insulin concentrations (Figure 27).
[00338] A 5:1 combination of Rho-isoalpha acids and Acacia was additionally tested in this model against metformin and roziglitazone, two pharmaceuticals currently in use for the treatment of diabetes. The results (Figure 28) indicate that the 5:1 combination of Rho- isoalpha acids and Acacia produced results compatible to the pharmaceutical agents in reducing serum glucose (panel A) and serum insulin (panel B).
[00339] The 2:1 and 5:1 combinations of Rho-isoalpha acids and Acacia appeared synergistic in the db/db murine diabetes model, supporting their potential usefulness in clinical situations requiring a reduction in serum glucose or enhance insulin sensitivity. Example 36
Effects of hops test compounds in a collagen induced rheumatoid arthritis murine model.
[00340] This example demonstrates the efficacy of two hops compounds, Mg Rho and
THIAA, in reducing inflamation and arthritic symptomology in a rheumatoid arthritis model, such inflammation and symptoms being known to mediated, in part, by a number of protein kinases.
[00341] The Model - Female DBA/J mice (10/group) were housed under standard conditions of light and darkness and allow diet ad libitum. The mice were injected intradermally on day 0 with a mixture containing 100 μg of type II collagen and 100 μg of Mycobacterium tuberculosis in squalene. A booster injection was repeated on day 21. Mice were examined on days 22 — 27 for arthritic signs with nonresponding mice removed from the study. Mice were treated daily by gavage with test compounds for 14 days beginning on day 28 and ending on day 42. Test compounds, as used in this example were RIAA (MgRho) at 10 mg/kg (lo), 50 mg/kg (med), or 250 mg/kg (hi); THIAA at 10 mg/kg (lo), 50 mg/kg (med), or 250 mg/kg (hi); celecoxib at 20 mg/kg; and prednisilone at 10 mg/kg.
[00342] Arthritic symptomology was assessed (scored 0 — 4) for each paw using a arthritic index as described below. Under this arthritic index 0 = no visible signs; 1 = edema and/or erythema: single digit; 2 = edema and or erythema: two joints; 3 = edema and or erythema: more than two joints; and 4 = severe arthritis of the entire paw and digits associated with ankylosis and deformity.
[00343] Histological examination^ At the termination of the experiment, mice were euthanized and one limb, was removed and preserved in buffered formalin. After the analysis of the arthritic index was found to be encouraging, two animals were selected at random from each treatment group for histological analysis by H&E staining. Soft tissue, joint and bone changes were monitored on a four point scale with a score of 4 indicating severe damage.
[00344] Cytokine analysis - Serum was collected from the mice at the termination of the experiment for cytokine analysis. The volume of sample being low (~ 0.2- 0.3 ml/mouse), samples from the ten mice were randomly allocated into two pools of five animals each. This was done so to permit repeat analyses; each analysis was performed a minimum of two times. TNF-ct and IL-6 were analyzed using mouse specific reagents (R&D Systems, Minneapolis, W
MN) according to the manufacturer's instructions. Only five of the twenty-six pools resulted in detectable levels of TNF-α; the vehicle treated control animal group was among them.
[00345] Results — The effect of RIAA on the arthritic index is presented graphically as
Figure 29. Significant reductions (p<0.05,two tail t-test) were observed for prednisolone at 10 mg/kg (days 30 - 42), celecoxib at 20 mg/kg (days 32 - 42), RIAA at 250 mg/kg (days 34 - 42) and RIAA at 50 mg/kg (days 38 - 40), demonstrating antiarthritc efficacy for RIAA at 50 or 250 mg/kg . Figure 30 displays the effects of THIAA on the arthritic index. Here, significant reductions were observed for celecoxib (days 32 — 42), THIAA at 250 mg/kg (days 34 — 42) and THIAA at 50 mg/kg (days 34 — 40), also demonstrating the effectiveness of THIAA as an antiarthritic agent.
[00346] The results from the histological examination of joint tissue damage are shown in Figure 31 and show the absence or minimal evidence of joint destruction in the THIAA treated individuals. There are clearly signs of a dose response and the reduction in the histology score at 250 mg/kg and 50 mg/ kg is 40% and 28% respectively. This compares favorably with the celecoxib treated group where joint destruction was scored as mild. Note that in the case of celecoxib (20 mg/kg) the histology score actually increased by 33%. There are obviously differences between individual animals, e.g. one of the vehicle treated animals showed evidence of moderate joint destruction while the other apparently free from damage. With the exception of one animal in the prednisolone treated group, synovitis was present in all treatment groups.
[00347] The results of the cytokine analysis for IL-6 are summarized in Figure 32.
With the exception of celecoxib, the high dose of Rho for all treatments reduced serum IL-6 levels, although only prednisolone reached a statistical significance.
Example 37
RIAA: Acacia (1:5) effects on metabolic syndrome in humans.
[00348] This experiment examined the effects treatment with a RIAA.Acacia (1:5) formulation on a number of clinically relevant markers in volunteer patients with metabolic syndrome. [00349] Methods and Trial Design - This trial was a randomized, placebo-controlled, double-blind trial conducted at a single study site (the Functional Medicine Research Center, Gig Harbor, WA). Inclusion criteria for the study required subjects (between 18 to 70 years of age) satisfy the following: (i) BMI between 25 and 42.5 kg/m2; (ii) TG/HDL-C ratio ≥3.5; (iii) fasting insulin ≥IO mcIU/mL. In addition, subjects had to meet 3 of the following 5 criteria: (i) waist circumference > 35" (women) and > 40" (men); (ii) TG >150 mg/dL; (iii) HDL < 50 mg/dL (women), and < 40 mg/dL (men); (iv) blood pressure > 130/85 or diagnosed hypertension on medication; and (v) fasting glucose Ξ≥IOO mg/dL.
[00350] Subjects who satisfied the inclusion criteria were randomized to one of 4 arms:
(i) subjects taking the KLAA/Acacia combination (containing 100 mg RIAA and 500 mg Acacia niloUca heartwood extract per tablet) at 1 tablet t.i.d.; (ii) subjects taking the KLAA/Acacia combination at 2 tablets t.i.d; (iii) placebo, 1 tablet, t.i.d; and (iv) placebo, 2 tablets, t.i.d. The total duration of the trial was 12 weeks. Blood was drawn from subjects at Day 1, at 8 weeks, and 12 weeks to assess the effect of supplementation on various parameters of metabolic syndrome.
[00351] Results - The initial demographic and biochemical characteristics of subjects
(pooled placebo group and subjects taking KLAA/Acacia at 3 tablets per day) enrolled for the trial are shown in Table 32. The initial fasting blood glucose and 2 h post-prandial (2 h pp) glucose values were similar between the KLAA/Acacia and placebo groups (99.0 vs. 96.5 mg/dL and 128.4 vs. 109.2 mg/dL, respectively). In addition, both glucose values were generally within the laboratory reference range (40-110 mg/dL for fasting blood glucose and 70-150 mg/dL for 2 h pp glucose). This was expected, because alteration in 2 h pp insulin response precedes the elevations in glucose and fasting insulin that are seen in later stage metabolic syndrome and frank diabetes. Table 32
Demographic and Baseline Biochemical Characteristics
Figure imgf000110_0001
*One subject was excluded from the analysis because of abnormal 2 h pp insulin values; BMI, Basal Metabolic Index; BP, Blood Pressure; TG, Triglyceride; HDL, High-Density Lipoprotein
[00352] Fasting blood insulin measurements were similar and generally within the reference range as well, with initial values of 17.5 mcIU/mL for the RIAA; 'Acacia group, and 13.2 mcIU/mL for the placebo group (reference range 3-30 mcIU/mL). The 2 h pp insulin levels were elevated past the reference range (99.3 vs. 80.2 mcIU/mL), and showed greater variability than did the fasting insulin or glucose measurements. Although the initial values were similar, the RIAA/ 'Acacia group showed a greater decrease in fasting insulin and 2 h pp insulin, as well as 2 h pp blood glucose after 8 weeks on the protocol (Figures 33 and 34).
[00353] The homeostatic model assessment (HOMA) score is a published measure of insulin resistance. The change in HOMA score for all subjects is shown in Figure 35. Due to the variability seen in metabolic syndrome subjects' insulin and glucose values, a subgroup of only those subjects with fasting insulin > 15 mcIU/mL was also assessed. The HOMA score for this subgroup is shown in Table 33, and indicates that a significant decrease was observed for the RIAA/Acacia group as compared to the placebo group.
Table 33
Effect of RIAA/Acacia supplementation (3 tablets/dav) on HOMA scores in subjects with initial fasting insulin > 15 mcIU/mL.
Figure imgf000111_0001
[00354] The difference between the groups was significant at 8 weeks (p < 0.05).
HOMA score was calculated from fasting insulin and glucose by published methods [(insulin (mcrU/mL)*glucose (mg/dL))/405].
[00355] Elevation in triglycerides (TG) is also an important suggestive indicator of metabolic syndrome. Table 34 and Figure 36 indicate that RIAA/Acacia supplementation resulted in a significant decrease in TG after 8 weeks as compared with placebo (p < 0.05). The TG/HDL-C ratio was also shown to decrease substantially for the RIAA/Acacia group (from 6.40 to 5.28), while no decrease was noted in the placebo group (from 5.81 to 5.92).
Table 34
Effect of RIAA/Acacia supplementation (3 tablets/dav) on TG levels and TG/HDL-
Cholesterol ratio.
Figure imgf000111_0002
[00356] Supplementation of metabolic syndrome subjects with a combination tablet composed of 100 rag rΛo-iso-alpha acids and 500 mg Acacia nilotica heartwood extract at 3 tablets per day for a duration of 8 weeks led to greater reduction of 2 h pp insulin levels, as compared to placebo. Further, greater decreases of fasting insulin, fasting and 2 h pp glucose, fasting triglyceride and HOMA scores were observed in subjects taking JUAA/Acacia supplement (3 tablets per day) versus subjects taking placebo. These results indicate RIAA/Λcacia supplementation might be useful in maintaining insulin homeostasis in subjects with metabolic syndrome.
Example 38
Effects of test compounds on cancer cell proliferation in vitro
[00357] This experiment demonstrates the direct inhibitory effects on cancer cell proliferation in vitro for a number of test compounds of the instant invention.
[00358] Methods - The colorectal cancer cell lines HT-29, Caco-2 and SW480 were seeded into 96-well plates at 3x103 cells/well and incubated overnight to allow cells to adhere to the plate. Each concentration of test material was replicated eight times. Seventy-two hours later, cells were assayed for total viable cells using the CyQUANT® Cell Proliferation Assay Kit. Percent decrease in viable cells relative to the DMSO solvent control was computed. Graphed values are means of eight observations ± 95% confidence intervals.
[00359] Results — Figures 37 — 41 graphically present the inhibitory effects of RIAA
(Fig. 37), IAA (Fig.38), THIAA (Fig.39), HHIAA (Fig. 40), and Xanthohumol (XN; Fig 41).
Example 39
Effects of celecoxib and test compounds on cancer cell proliferation in vitro
[00360] This experiment compares the observed versus expected inhibitory effects on cancer cell proliferation in vitro of RIAA or THIAA in combination with celecoxib.
[00361] Methods - The colorectal cancer cell lines were seeded into 96-well plates at
3x103 cells/well and incubated overnight to allow cells to adhere to the plate. Each concentration of test material was replicated eight times. Seventy-two hours later, cells were assayed for total viable cells using the CyQUANT® Cell Proliferation Assay Kit. The OBSERVED percent decrease in viable cells relative to the DMSO solvent control was computed. Estimates of the EXPECTED cytotoxic effect of celecoxib and RIAA or THIAA combinations were made using the relationship: l/[T]c = X/[T]x + Y/[T]y, where T = the toxicity represented as fraction of the growth inhibited or cells killed, X and Y are the relative fractions of each component in the test mixture, and X + Y = 1. Graphed OBSERVED values are means of eight observations ± 95% confidence intervals. Synergy was inferred when the ESTIMATED percent decrease fell below the 95% confidence interval of the corresponding OBSERVED fraction.
[00362] Figures 42 and 43 graphically present a comparison between the observed and expected inhibitory effects of RIAA (Fig. 42) or THIAA (Fig. 43) on cancer cell proliferation. These results indicate that the compounds tested in combination with celecoxib inhibited cancer cell proliferation to an extent greater than mathematically predicted in most instances.
Example 40
Detection of THIAA in Serum Following Oral Dosage
[00363] The purpose of this experiment was to determine whether THIAA was metabolized and detectable following oral dosage.
[00364] Methods — Following a predose blood draw, five softgels (188mg THIAA/ softgel) delivering 940 mg of THIAA as the free acid (PR Tetra Standalone Softgel. OG# 2210 KP-247. Lot C42331111) were consumed and immediately followed by a container of fruit yogurt. With the excpetion of decaffinated coffee, no additional food was consumed over the next four hours following THIAA ingestion. Samples were drawn at 45 minute intervals into Corvac Serum Separator tubes with no clot activator. Samples were allowed to clot at room temperature for 45 minutes and serum separated by centrifugation at 1800 x g for 10 minutes at 4° C. To 0.3 ml of serum 0.9 ml of MeCN containing 0.5% HOAc was added and kept at -20° C for 45-90 minutes. The mixture was centrifuged at 15000 x g for 10 minutes at 4°C. Two phases were evident following centrifugation two phases were evident; 0.6 ml of the upper phase was sampled for HPLC analysis. Recovery was determined by using spiked samples and was greater than 95%.
Ill [00365] Results - The results are presented graphically as Figures 44 - 46. Figure 44 graphically displays the detection of THIAA in the serum over time following ingestion of 940 mg of THIAA. Figure 45 demonstrates that after 225 minutes following ingestion, THIAA was detected in the serum at levels comparabe to those THIAA levels tested in vitro. Figure 46 depicts the metabolism of THIAA by CYP2C9*1.
[00366] The invention now having been fully described, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto without departing from the spirit or scope of the appended claims.

Claims

1. A method to treat a cancer responsive to protein kinase modulation in a mammal in need thereof, said method comprising administering to the mammal a therapeutically effective amount of a hexahydro-isoalpha acid.
2. The method of Claim 1, wherein the hexahydro-isoalpha acid is selected from the group consisting of hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.
3. The method of Claim 1, wherein the protein kinase modulated is selected from the group consisting of Abl(T315I), Aurora-A, Bmx, CDK9/cyclin Tl, CKl γl, CKI72, CKI73, cSRC, DAPKl, DAPK2, EphBl, ErbB4, Fer, FGFR2, GSK3B, GSK3α, HIPK3, IGF-IR, MAPKAP-K2, MSK2, PAK3, PAK5, PI3K, Pim-1, PKA(b), PKBB, PKBγ, PRAK, Rsk2, Syk, Tie2, TrkA, TrkB, and ZIPK.
4. The method of Claim 1, wherein the cancer responsive to kinase modulation is selected from the group consisting of bladder, breast, cervical, colon, lung, lymphoma, melanoma, prostate, thyroid, and uterine cancer.
5. A composition to treat a cancer responsive to protein kinase modulation in a mammal in need thereof, said composition comprising a therapeutically effective amount of a hexahydro-isoalpha acid; wherein said therapeutically effective amount modulates a cancer associated protein kinase.
6. The composition of Claim 5, wherein the hexahydro-isoalpha acid is selected from the group consisting of hexahydro-isohumulone, hexahydro-isocohumulone, and hexahydro-adhumulone.
7. The composition of Claim 5, wherein the composition further comprises a pharmaceutically acceptable excipient selected from the group consisting of coatings, isotonic and absorption delaying agents, binders, adhesives, lubricants, disintergrants, coloring agents, flavoring agents, sweetening agents, absorbants, detergents, and emulsifying agents. The composition of Claim 5, wherein the composition further comprises one or more members selected from the group consisting of antioxidants, vitamins, minerals, proteins, fats, and carbohydrates.
PCT/US2007/014450 2006-06-20 2007-06-20 Hexahydro-isoalpha acid based protein kinase modulation cancer treatment WO2007149523A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2009516569A JP2009541329A (en) 2006-06-20 2007-06-20 Protein kinase-regulated cancer therapy based on hexahydro-isoalpha acid
AU2007261356A AU2007261356A1 (en) 2006-06-20 2007-06-20 Hexahydro-isoalpha acid based protein kinase modulation cancer treatment
CA002655059A CA2655059A1 (en) 2006-06-20 2007-06-20 Hexahydro-isoalpha acid based protein kinase modulation cancer treatment
EP07796314A EP2043621A4 (en) 2006-06-20 2007-06-20 Hexahydro-isoalpha acid based protein kinase modulation cancer treatment

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81506406P 2006-06-20 2006-06-20
US60/815,064 2006-06-20

Publications (2)

Publication Number Publication Date
WO2007149523A2 true WO2007149523A2 (en) 2007-12-27
WO2007149523A3 WO2007149523A3 (en) 2008-09-04

Family

ID=38833737

Family Applications (8)

Application Number Title Priority Date Filing Date
PCT/US2007/014380 WO2007149485A1 (en) 2006-06-20 2007-06-20 Acacia based protein kinase modulation cancer treatment
PCT/US2007/014412 WO2007149503A2 (en) 2006-06-20 2007-06-20 Xanthohumol and tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014372 WO2007149480A2 (en) 2006-06-20 2007-06-20 Reduced isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014413 WO2007149504A2 (en) 2006-06-20 2007-06-20 Isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014374 WO2007149482A2 (en) 2006-06-20 2007-06-20 Xanthohumol based protein kinase modulation cancer treatment
PCT/US2007/014450 WO2007149523A2 (en) 2006-06-20 2007-06-20 Hexahydro-isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014373 WO2007149481A2 (en) 2006-06-20 2007-06-20 Tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014414 WO2007149505A2 (en) 2006-06-20 2007-06-20 Beta acid based protein kinase modulation cancer treatment

Family Applications Before (5)

Application Number Title Priority Date Filing Date
PCT/US2007/014380 WO2007149485A1 (en) 2006-06-20 2007-06-20 Acacia based protein kinase modulation cancer treatment
PCT/US2007/014412 WO2007149503A2 (en) 2006-06-20 2007-06-20 Xanthohumol and tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014372 WO2007149480A2 (en) 2006-06-20 2007-06-20 Reduced isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014413 WO2007149504A2 (en) 2006-06-20 2007-06-20 Isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014374 WO2007149482A2 (en) 2006-06-20 2007-06-20 Xanthohumol based protein kinase modulation cancer treatment

Family Applications After (2)

Application Number Title Priority Date Filing Date
PCT/US2007/014373 WO2007149481A2 (en) 2006-06-20 2007-06-20 Tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
PCT/US2007/014414 WO2007149505A2 (en) 2006-06-20 2007-06-20 Beta acid based protein kinase modulation cancer treatment

Country Status (9)

Country Link
US (6) US20080031894A1 (en)
EP (4) EP2043622A4 (en)
JP (4) JP2009541326A (en)
KR (4) KR20090026191A (en)
CN (4) CN101573128A (en)
AU (4) AU2007261399A1 (en)
CA (4) CA2655047A1 (en)
TW (8) TW200819121A (en)
WO (8) WO2007149485A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106153920A (en) * 2016-07-25 2016-11-23 四川大学华西医院 A kind of screening lung cancer test kit

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7736677B2 (en) * 2001-06-20 2010-06-15 Metaproteomics, Llc Xanthohumol and tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
CN101573128A (en) * 2006-06-20 2009-11-04 麦特普罗泰欧米克斯有限公司 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
EP2052728B1 (en) * 2006-08-10 2018-06-13 mimozax Co., Ltd. Hypoglycemic composition containing component originating in the bark of tree belonging to the genus acacia
KR101398737B1 (en) * 2006-08-10 2014-05-27 가부시키가이샤 미모잭스 Composition for preventing and/or treating tumor containing component originating in the bark of tree belonging to the genus acacia
US20100160450A1 (en) * 2007-01-31 2010-06-24 Eric Kuhrts Methods of reducing 15-f2t-isop levels in mammals
MX2010006425A (en) * 2007-12-10 2010-08-31 Metaproteomics Llc Substituted 1,3-cyclopentadione multi-target protein kinase modulators of cancer, angiogenesis and the inflammatory pathways associated therewith.
US20110092591A1 (en) * 2008-02-06 2011-04-21 Noscira S.A. Phenyl-prenyl derivatives, of marine and synthetic origin, for the treatment of cognitive, neurodegenerative or neuronal diseases or disorders
JP2010043064A (en) * 2008-07-16 2010-02-25 Sapporo Breweries Ltd Fat cell differentiation inhibitor
WO2010056406A1 (en) 2008-11-12 2010-05-20 The United State Of America, As Represented By The Secretary, Department Of Health & Human Services Use of erbb4 as a prognostic and therapeutic marker for melanoma
AU2010300531A1 (en) 2009-09-30 2012-05-24 President And Fellows Of Harvard College Methods for modulation of autophagy through the modulation of autophagy-inhibiting gene products
CN102724994A (en) * 2010-01-11 2012-10-10 希尔洛有限公司 Method for treatment of inflammatory disease and disorder
WO2011163466A1 (en) 2010-06-23 2011-12-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (nrg-1)
EP2720693B8 (en) 2011-06-17 2017-05-17 Ludwig Aigner Chromane-like cyclic prenylflavonoids for the medical intervention in neurological disorders
US9499856B2 (en) 2012-04-02 2016-11-22 The Board Institute, Inc. DDR2 mutations in squamous cell lung cancer
CN104399044A (en) * 2014-12-01 2015-03-11 郑州后羿制药有限公司 Traditional Chinese veterinary medicine for treating arthritis, rheumatoid arthritis and osteoproliferation
CN105168946A (en) * 2015-10-22 2015-12-23 陈远征 Traditional Chinese medicine composition for treating diabetes mellitus and application thereof
CN105126040A (en) * 2015-10-23 2015-12-09 戚炎月 Medicine compound treating ovarian cysts and preparation method thereof
US10918650B2 (en) * 2016-06-02 2021-02-16 University Of South Florida Method of treating melanoma using an inhibitor of an atypical protein kinase C
CN107115328B (en) * 2017-05-24 2019-08-30 中美(河南)荷美尔肿瘤研究院 Application of the xanthohumol in terms of preparing protein kinase B inhibitor
CN108535480B (en) * 2018-03-05 2020-03-06 南通大学附属医院 Application of EphA8 gene in preparation of anti-breast cancer drug and diagnostic kit thereof
CN108586226B (en) * 2018-05-31 2021-06-18 温州医科大学 3-methyl-3-butene-2-alcohol chalcone compound and synthesis and application thereof
CN110833550B (en) * 2018-08-15 2023-03-24 广西梧州制药(集团)股份有限公司 Application of pyrazolopyrimidine derivative in treatment of liver injury caused by acute pancreatitis
CN114921546B (en) * 2022-05-13 2023-02-21 核工业总医院 Application of circHIPK2 as breast cancer biomarker
CN116102416A (en) * 2023-02-21 2023-05-12 蚌埠医学院 Bavachin derivatives, preparation method thereof and application thereof in preparation of anticancer drugs
CN116196301B (en) * 2023-04-27 2023-07-28 北京中医药大学 Chalcone alpha-glucosidase inhibitor and preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040115290A1 (en) 2001-06-20 2004-06-17 Tripp Matthew L. Modulation of inflammation by hops fractions and derivatives
US20040151792A1 (en) 2001-06-20 2004-08-05 Tripp Matthew L. Compositions that treat or inhibit pathological conditions associated with inflammatory response

Family Cites Families (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3933919A (en) * 1964-12-15 1976-01-20 Geoffrey Wilkinson Hydroformylation of mono-α-olefins and mono-α-acetylenes
US3451921A (en) * 1965-01-25 1969-06-24 Union Carbide Corp Coke production
GB1145240A (en) * 1965-03-01 1969-03-12 Kalamazoo Spice Extract Co Hop flavours for malt beverages and the like
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
US3536495A (en) * 1968-03-13 1970-10-27 Miller Brewing Ammonia complexes of hop alpha acids and modified alpha acids
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
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
JPS52145509A (en) * 1976-05-27 1977-12-03 Tokutarou Matsui Antitumor agent
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
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
US4590296A (en) * 1984-01-25 1986-05-20 Miller Brewing Company Process for separation of beta-acids from extract containing alpha-acids and beta-acids
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
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
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
AU4715093A (en) * 1992-07-29 1994-03-03 Matforsk, Norwegian Food Research Institute Composition comprising fertilized shell eggs
US5286506A (en) * 1992-10-29 1994-02-15 Bio-Technical Resources Inhibition of food pathogens by hop acids
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
EP0992581B2 (en) * 1993-11-04 2011-05-25 Innogenetics N.V. Immunodominant human T-cell epitopes of hepatitis C virus
JP2677762B2 (en) * 1994-04-08 1997-11-17 株式会社神戸製鋼所 Oil-cooled compressor
DE69507185T2 (en) * 1994-04-12 1999-09-09 Hoechst Marion Roussel Inc Medicines used to treat osteoporosis
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
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
US7045519B2 (en) * 1998-06-19 2006-05-16 Chiron Corporation Inhibitors of glycogen synthase kinase 3
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.
US6801860B1 (en) * 1999-02-15 2004-10-05 Genetics Institute, Llc Crystal structure of cPLA2 and methods of identifying agonists and antagonists using same
US6462029B1 (en) * 1999-02-23 2002-10-08 Econugenics Compositions and methods for treating mammals with modified alginates and modified pectins
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
US6129907A (en) * 1999-08-04 2000-10-10 Colgate Palmolive Company Stable hydrogenated lupulone antibacterial oral compositions
AU7596100A (en) * 1999-09-21 2001-04-24 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
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
EP1295587A1 (en) * 2000-03-31 2003-03-26 The Nisshin OilliO, Ltd. External preparation for the 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
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
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
US7078062B2 (en) * 2001-01-17 2006-07-18 S.S. Steiner, Inc. Hop-based udder and teat dips and washes
US20020187239A1 (en) * 2001-02-06 2002-12-12 Dusan Miljkovic Nutraceuticals and methods of obtaining nutraceuticals from tropical crops
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
US8142819B2 (en) * 2002-10-21 2012-03-27 Metaproteomics, Llc Synergistic 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
US7736677B2 (en) * 2001-06-20 2010-06-15 Metaproteomics, Llc Xanthohumol and tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
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
US8168234B2 (en) * 2001-06-20 2012-05-01 Metaproteomics, Llc Compositions that treat or inhibit pathological conditions associated with inflammatory response
US7205151B2 (en) * 2001-06-20 2007-04-17 Metaproteomics, Llc Complex mixtures exhibiting selective inhibition of cyclooxygenase-2
US20030082511A1 (en) * 2001-09-25 2003-05-01 Brown Steven J. Identification of modulatory molecules using inducible promoters
MXPA04003734A (en) * 2001-10-26 2005-06-20 Metaproteomics Llc 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
ATE361746T1 (en) * 2002-03-06 2007-06-15 Medical Res And Education Trus BOTANICAL EXTRACT WITH ANTI-CANCER ACTIVITY CONTAINING ISOLIQUIRITIGENIN
WO2004037180A2 (en) * 2002-10-21 2004-05-06 Metaproteomics, Llc Compositions that treat or inhibit pathological conditions associated with inflammatory response
US7144590B2 (en) * 2003-01-09 2006-12-05 Lipoprotein Technologies, Inc. Bioactive compositions derived from humulus lupulus
MXPA05013061A (en) * 2003-06-05 2006-03-02 Warner Lambert Co Cycloalkyl and heterocycloalkyl substituted benzothiophenes as therapeutic agents.
GB0317020D0 (en) * 2003-07-21 2003-08-27 Sahajanand Biotech Private Ltd Herbo-mineral formulation for refractory leukemias and lymphomas
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
US20090274778A1 (en) * 2004-11-13 2009-11-05 Babish John G Compositions Exhibiting Inhibition Of Cyclooxygenase-2
MX2008001903A (en) * 2005-08-09 2008-04-16 Metaproteomics Llc Protein kinase modulation by hops and acacia products.
US20070065456A1 (en) * 2005-09-20 2007-03-22 Woods Cindy J Nutritional supplements
US20070154576A1 (en) * 2005-12-09 2007-07-05 Tripp Matthew L Protein kinase modulation by hops and Acacia products
CN101573128A (en) * 2006-06-20 2009-11-04 麦特普罗泰欧米克斯有限公司 Tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
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.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040115290A1 (en) 2001-06-20 2004-06-17 Tripp Matthew L. Modulation of inflammation by hops fractions and derivatives
US20040151792A1 (en) 2001-06-20 2004-08-05 Tripp Matthew L. Compositions that treat or inhibit pathological conditions associated with inflammatory response

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2043621A4

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106153920A (en) * 2016-07-25 2016-11-23 四川大学华西医院 A kind of screening lung cancer test kit
CN106153920B (en) * 2016-07-25 2018-04-27 四川大学华西医院 A kind of screening lung cancer kit

Also Published As

Publication number Publication date
KR20090023722A (en) 2009-03-05
CA2655047A1 (en) 2007-12-27
JP2009541326A (en) 2009-11-26
WO2007149481A2 (en) 2007-12-27
JP2009541324A (en) 2009-11-26
WO2007149480A3 (en) 2008-07-10
WO2007149505A2 (en) 2007-12-27
EP2043622A2 (en) 2009-04-08
KR20090023719A (en) 2009-03-05
WO2007149523A3 (en) 2008-09-04
US20080033056A1 (en) 2008-02-07
WO2007149503A2 (en) 2007-12-27
JP2009541329A (en) 2009-11-26
CA2655043A1 (en) 2007-12-27
WO2007149504A3 (en) 2008-03-06
TW200817022A (en) 2008-04-16
TW200816980A (en) 2008-04-16
TW200817026A (en) 2008-04-16
AU2007261400A1 (en) 2007-12-27
CA2655059A1 (en) 2007-12-27
EP2046353A4 (en) 2010-01-27
EP2046353A2 (en) 2009-04-15
US20080033057A1 (en) 2008-02-07
EP2043621A2 (en) 2009-04-08
KR20090023721A (en) 2009-03-05
CA2654964A1 (en) 2007-12-27
EP2043622A4 (en) 2010-02-24
CN101505770A (en) 2009-08-12
WO2007149504A2 (en) 2007-12-27
CN101505742A (en) 2009-08-12
JP2009541325A (en) 2009-11-26
WO2007149480A2 (en) 2007-12-27
TW200817023A (en) 2008-04-16
EP2043621A4 (en) 2009-08-26
CN101573128A (en) 2009-11-04
AU2007261399A1 (en) 2007-12-27
TW200817027A (en) 2008-04-16
US20080026088A1 (en) 2008-01-31
EP2046355A2 (en) 2009-04-15
WO2007149482A2 (en) 2007-12-27
WO2007149505A3 (en) 2008-05-02
WO2007149485A1 (en) 2007-12-27
CN101505743A (en) 2009-08-12
EP2046355A4 (en) 2010-02-03
WO2007149481A3 (en) 2008-11-27
TW200819120A (en) 2008-05-01
TW200816982A (en) 2008-04-16
US20080031894A1 (en) 2008-02-07
AU2007261356A1 (en) 2007-12-27
WO2007149482A3 (en) 2008-05-08
KR20090026191A (en) 2009-03-11
TW200819121A (en) 2008-05-01
US20080031982A1 (en) 2008-02-07
WO2007149503A3 (en) 2008-05-02
AU2007261338A1 (en) 2007-12-27
US20080031893A1 (en) 2008-02-07

Similar Documents

Publication Publication Date Title
US8263139B2 (en) Protein kinase modulation by hops and Acacia products
US7736677B2 (en) Xanthohumol and tetrahydro-isoalpha acid based protein kinase modulation cancer treatment
US20070154576A1 (en) Protein kinase modulation by hops and Acacia products
WO2007149523A2 (en) Hexahydro-isoalpha acid based protein kinase modulation cancer treatment
AU2012204133B2 (en) Protein kinase modulation by hops and acacia products
US20080051466A1 (en) Isoalpha acid based protein kinase modulation cancer treatment
AU2013202154A1 (en) Beta acid based protein kinase modulation cancer treatment
AU2013200576A1 (en) Protein kinase modulation by hops and Acacia products
MX2008007273A (en) Protein kinase modulation by hops and acacia products

Legal Events

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

Ref document number: 200780030611.9

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07796314

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2655059

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009516569

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007261356

Country of ref document: AU

NENP Non-entry into the national phase

Ref country code: RU

WWE Wipo information: entry into national phase

Ref document number: 1020097001249

Country of ref document: KR

Ref document number: 2007796314

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2007261356

Country of ref document: AU

Date of ref document: 20070620

Kind code of ref document: A