WO2004046345A2 - Use of hmgb fragments as anti-inflammatory agents - Google Patents

Use of hmgb fragments as anti-inflammatory agents Download PDF

Info

Publication number
WO2004046345A2
WO2004046345A2 PCT/US2003/037507 US0337507W WO2004046345A2 WO 2004046345 A2 WO2004046345 A2 WO 2004046345A2 US 0337507 W US0337507 W US 0337507W WO 2004046345 A2 WO2004046345 A2 WO 2004046345A2
Authority
WO
WIPO (PCT)
Prior art keywords
box
hmgb
polypeptide
fragment
group
Prior art date
Application number
PCT/US2003/037507
Other languages
French (fr)
Other versions
WO2004046345A3 (en
Inventor
Walter Newman
Theresa L. O'keefe
Original Assignee
Critical Therapeutics, Inc.
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 Critical Therapeutics, Inc. filed Critical Critical Therapeutics, Inc.
Priority to CA002506328A priority Critical patent/CA2506328A1/en
Priority to AU2003294488A priority patent/AU2003294488B2/en
Priority to NZ540067A priority patent/NZ540067A/en
Priority to EP03789973A priority patent/EP1569684A4/en
Priority to JP2004554020A priority patent/JP2006510619A/en
Publication of WO2004046345A2 publication Critical patent/WO2004046345A2/en
Publication of WO2004046345A3 publication Critical patent/WO2004046345A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives

Definitions

  • Inflammation is often induced by proinflammatory cytokines, such as tumor necrosis factor (TNF), inter leukin (IL)-l , IL-l ⁇ , IL-6, platelet-activating factor (PAF), macrophage migration inhibitory factor (MIF), and other compounds.
  • TNF tumor necrosis factor
  • IL inter leukin
  • PAF platelet-activating factor
  • MIF macrophage migration inhibitory factor
  • proinflammatory cytokines are produced by several different cell types, most importantly immune cells (for example, monocytes, macrophages and neutrophils), but also non-immune cells such as f ⁇ broblasts, osteoblasts, smooth muscle cells, epithelial cells, and neurons.
  • TNF tumor necrosis factor
  • IL-l inter leukin
  • IL-6 platelet-activating factor
  • MIF macrophage migration inhibitory factor
  • proinflammatory cytokines are produced by several different cell types, most importantly immune cells (for example, monocytes, macrophages
  • Inflammatory cytokine cascades contribute to deleterious characteristics, including inflammation and apoptosis, of numerous disorders. Included are disorders characterized by both localized and systemic reactions, including, without limitation, diseases involving the gastrointestinal tract and associated tissues (such as appendicitis, peptic, gastric and duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute and ischemic colitis, diverticulitis, epiglottitis, achalasia, cholangitis, cholecystitis, coeliac disease, hepatitis, Crohn's disease, enteritis, and Whipple's disease); systemic or local inflammatory diseases and conditions (such as asthma, allergy, anaphylactic shock, immune complex disease, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, hyperpyrexia, eosinophilic granuloma
  • HMGB1 high mobility group box 1
  • HMG-1 and HMG1 a protein that accumulates in serum and mediates delayed lethality and further induction of early proinflammatory cytokines.
  • HMGBl was first identified as the founding member of a family of DNA- binding proteins termed high mobility group box (HMGB) proteins that are critical for DNA structure and stability. It was identified nearly 40 years ago as a ubiquitously expressed nuclear protein that binds double-stranded DNA without sequence specificity.
  • HMGBl binding bends DNA to promote formation and stability of nucleoprotein complexes that facilitate gene transcription of glucocorticoid receptors and RAG recombinase.
  • the HMGBl molecule has three domains: two DNA binding motifs termed HMGB A and HMGB B boxes, and an acidic carboxyl terminus.
  • the two HMGB boxes are highly conserved 80 amino acid, L-shaped domains.
  • HMGB boxes are also expressed in other transcription factors including the RNA polymerase I transcription factor human upstream-binding factor and lymphoid-specific factor.
  • HMGBl As a cytokine mediator of inflammatory conditions.
  • HMGBl has been implicated as a cytokine mediator of delayed lethality in endotoxemia. That work demonstrated that bacterial endotoxin (lipopolysaccharide (LPS)) activates monocytes/macrophages to release HMGBl as a late response to activation, resulting in elevated serum HMGBl levels that are toxic.
  • LPS lipopolysaccharide
  • Antibodies against HMGB 1 prevent lethality of endotoxin even when antibody administration is delayed until after the early cytokine response.
  • HMGBl is a potent activator of monocytes.
  • HMGBl Intratracheal application of HMGBl causes acute lung injury, and anti-HMGBl antibodies protect against endotoxin-induced lung edema. Serum HMGBl levels are elevated in critically ill patients with sepsis or hemorrhagic shock, and levels are significantly higher in non-survivors as compared to survivors.
  • HMGBl has also been implicated as a ligand for RAGE, a multi-ligand receptor ofthe immunoglobulin superfamily.
  • RAGE is expressed on endothelial cells, smooth muscle cells, monocytes, and nerves, and ligand interaction transduces signals through MAP kinase, P21 ras, andNF- ⁇ B.
  • the present invention is based on the discoveries that (1) the HMGB A box serves as a competitive inhibitor of HMGB proinflammatory action, (2) the HMGB B box has the predominant proinflammatory activity of HMGB, and (3) combination therapies involving agents that inhibit HMGB biological activity and agents that inhibit TNF biological activity can be used for the treatment of conditions characterized by activation ofthe inflammatory cytokine cascade.
  • Agents that inhibit HMGB biological activity include the HMGB A box, which serves as a competitive inhibitor of HMGB proinflammatory action, and antibodies to HMGB, for example, the HMGB B box.
  • the invention is a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof, or an A box biologically active fragment or variant thereof, which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, wherein the HMGB A box is selected from the group consisting of an
  • HMG1L5 (formerly HMG1L10) A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RP11-395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box.
  • the polypeptide can be in a pharmaceutically acceptable carrier.
  • the invention is a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, wherein the antibodies can inhibit release of a proinflammatory cytokine from a cell treated with HMGB, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23.
  • the antibodies can be in a pharmaceutically acceptable carrier.
  • the invention is a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, or a B box biologically active fragment or variant thereof, but not comprising a full length HMGB, wherein the polypeptide can cause release of a proinflammatory cytokine from a cell, and wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23.
  • the polypeptide can be in a pharmaceutically acceptable carrier.
  • the invention comprises vectors encoding the polypeptides described above.
  • the invention is a method of inhibiting release of a proinflammatory cytokine from a mammalian cell, the method comprising treating the cell with an amount of a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (fonnerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23.
  • HMGB high mobility group box protein
  • the invention is a method of inhibiting release of a proinflammatory cytokine from a mammalian cell, the method comprising treating the cell with a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof, or an A box biologically active fragment or variant thereof, which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in an amount sufficient to inhibit release ofthe proinflammatory cytokine from the cell, wherein the HMGB A box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RP11- 395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box.
  • the cell can be treated with a vector encoding a polypeptide
  • the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, in an amount sufficient to inhibit the inflammatory cytokine cascade, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
  • HMG1L5 originally HMG1L10
  • the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof, or an A box biologically active fragment or variant thereof, which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, in an amount sufficient to inhibit release ofthe proinflammatory cytokine from the cell, wherein the HMGB A box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl l-395A23, an HMGlL9 A box, an LOC122441 B box, an LOCI 39603 A box, and an HMG1L8 A box.
  • HMGB high mobility group box protein
  • the invention is a method of stimulating the release of a proinflammatory cytokine from a cell comprising treating the cell with a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, or a B box biologically active fragment thereof, but not comprising a full length HMGB, in an amount sufficient to stimulate the release ofthe proinflammatory cytokine from the cell, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1- 395A23.
  • the cell can be treated with a vector encoding a polypeptide comprising the B box polypeptide, B box biologically active fragment, or variant thereof.
  • the invention is a method for effecting weight loss or treating obesity in a patient, comprising administering to the patient an effective amount of a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, or a B box biologically active fragment or variant thereof, but not comprising a full length HMGB polypeptide, in an amount sufficient to stimulate the release of a proinflammatory cytokine from a cell, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
  • HMGB high mobility group box protein
  • the invention is a method of determining whether a compound inhibits inflammation, comprising combining the compound with a) a cell that releases a proinflammatory cytokine when exposed to a high mobility group box protein (HMGB) B box or a biologically active fragment thereof; and b) the HMGB B box or biologically active fragment thereof, wherein said HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23; then determining whether the compound inhibits the release of the proinflammatory cytokine from the cell.
  • HMGB high mobility group box protein
  • the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a polypeptide comprising a high mobility group box (HMGB) A box, or a fragment or variant thereof, that can inhibit release of a proinflammatory cytokine from a cell treated with a high mobility group box (HMGB) protein and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide, in a pharmaceutically acceptable carrier.
  • HMGB high mobility group box
  • the HMGB A box is preferably a vertebrate HMGB A box, for example, a mammalian HMGB A box, more preferably, a mammalian HMGBl A box, for example, a human HMGBl A box, and most preferably, the HMGBl A box comprising or consisting ofthe sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57.
  • the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody that binds an HMGB polypeptide or a biologically active fragment thereof, for example, an HMGB B box polypeptide or biologically active fragment thereof, and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab,
  • CDP870 CDP571, Lenercept, and Thalidomide, in a pharmaceutically acceptable carrier.
  • the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade comprising administering to the patient a composition comprising a polypeptide comprising a high mobility group box (HMGB) A box or a fragment or variant thereof that can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide.
  • HMGB high mobility group box
  • the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade comprising administering to the patient a composition comprising an antibody that binds an HMGB polypeptide or a biologically active fragment thereof, for example, an HMGB B box polypeptide or a biologically active fragment thereof, and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide.
  • FIG. 1 is a schematic representation of HMGBl mutants and their activity in TNF release (pg/ml).
  • FIG. 2A is a histogram showing the effect of 0 ⁇ g/ml, 0.01 ⁇ g/ml, 0.1 ⁇ g/ml, 1 ⁇ g/ml or 10 ⁇ g/ml of B box on TNF release (pg/ml) in RAW 264.7 cells.
  • FIG. 2B is a histogram showing the effect of 0 ⁇ g/ml, 0.01 ⁇ g/ml, 0.1 ⁇ g/ml, 1 ⁇ g/ml or 10 ⁇ g/ml of B box on IL-l ⁇ release (pg/ml) in RAW 264.7 cells.
  • FIG. 2C is a histogram showing the effect of 0 ⁇ g/ml, 0.01 ⁇ g/ml, 0.1 ⁇ g/ml, 1 ⁇ g/ml or 10 ⁇ g/ml of B box on IL-6 release (pg/ml) in RAW 264.7 cells.
  • FIG. 2D a scanned image of a blot of an RNAse protection assay, showing the effect of B box (at 0 hours, 4 hours, 8 hours, or 24 hours after administration) or vector alone (at 4 hours after administration) on TNF mRNA expression in RAW 264.7 cells.
  • FIG. 2E is a histogram ofthe effect of HMGBl B box on TNF protein release (pg/ml) from RAW 264.7 cells at 0 hours, 4 hours, 8 hours, 24 hours, 32 hours or 48 hours after administration.
  • FIG. 2F is a histogram ofthe effect of vector on TNF protein release (pg/ml) from RAW 264.7 cells at 0 hours, 4 hours, 8 hours, 24 hours, 32 hours or 48 hours after administration.
  • FIG. 3 is a schematic representation of HMGB 1 B box mutants and their activity in TNF release (pg/ml).
  • FIG. 4A is a graph ofthe effect of 0 ⁇ g/ml, 5 ⁇ g/ml, 10 ⁇ g/ml, or 25 ⁇ g/ml of HMG1 A box protein on the release of TNF (as a percent of HMGBl mediated TNF release alone) from RAW 264.7 cells.
  • FIG. 4B is a histogram of the effect of HMGB 1 (0 or 1.5 ⁇ g/ml), HMGB 1 A box (0 or 10 ⁇ g/ml), or vector (0 or 10 ⁇ g/ml), alone, or in combination, on the release of TNF (as a percent of HMGBl mediated TNF release alone) from RAW 264.7 cells.
  • FIG. 5A is a graph of binding of 125 I-HMGB1 binding to RAW 264.7 cells (CPM/well) over time (minutes).
  • FIG. 5B is a histogram ofthe binding of 125 I-HMGB1 in the absence of unlabeled HMGBl or HMGBl A box for 2 hours at 4°C (Total), or in the presence of 5,000 molar excess of unlabeled HMGBl (HMGBl) or A box (A box), measured as a percent ofthe total CPM/well.
  • FIG. 5B is a histogram ofthe binding of 125 I-HMGB1 in the absence of unlabeled HMGBl or HMGBl A box for 2 hours at 4°C (Total), or in the presence of 5,000 molar excess of unlabeled HMGBl (HMGBl) or A box (A box), measured as a percent ofthe total CPM/well.
  • HMGB 1 HMG-1 ; 0 ⁇ g/ml or 1 ⁇ g/ml
  • HMGBl B box B Box; 0 ⁇ g/ml or 10 ⁇ g/ml
  • anti-B box antibody 25 ⁇ g/ml or 100 ⁇ g/ml
  • IgG 25 ⁇ g/ml or 100 ⁇ g/ml
  • FIG. 7A is a scanned image of a hematoxylin and eosin stained kidney section obtained from an untreated mouse.
  • FIG. 7B is a scanned image of a hematoxylin and eosin stained kidney section obtained from a mouse administered HMGBl B box.
  • FIG. 7C is a scanned image of a hematoxylin and eosin stained myocardium section obtained from an untreated mouse.
  • FIG. 7D is a scanned image of a hematoxylin and eosin stained myocardium section obtained from a mouse administered HMGBl B box.
  • FIG. 7E is a scanned image of a hematoxylin and eosin stained lung section obtained from an untreated mouse.
  • FIG. 7F is a scanned image of a hematoxylin and eosin stained lung section obtained from a mouse administered HMGB 1 B box.
  • FIG. 7G is a scanned image of a hematoxylin and eosin stained liver section obtained from an untreated mouse.
  • FIG. 7H is a scanned image of a hematoxylin and eosin stained liver section obtained from a mouse administered HMGBl B box.
  • FIG. 71 is a scanned image of a hematoxylin and eosin stained liver section
  • FIG. 7J is a scanned image of a hematoxylin and eosin stained liver section (high magnification) obtained from a mouse administered HMGBl B box.
  • FIG. 8 is a graph ofthe level of HMGBl (ng/ml) in mice subjected to cecal ligation and puncture (CLP) over time (hours).
  • FIG. 9 is a graph ofthe effect of HMGB A Box (60 ⁇ g/mouse or 600 ⁇ g/mouse) or no treatment on survival of mice over time (days) after cecal ligation and puncture (CLP).
  • FIG. 10A is a graph ofthe effect of anti-HMGBl antibody (dark circles) or no treatment (open circles) on survival of mice over time (days) after cecal ligation and puncture (CLP).
  • FIG. 1 OB is a graph ofthe effect of anti-HMGBl B box antiserum ( ⁇ ) or no treatment (*) on the survival (days) of mice administered lipopolysaccharide (LPS).
  • FIG. 11A is a histogram ofthe effect of anti-RAGE antibody or non-immune IgG on TNF release from RAW 264.7 cells treated with HMGBl (HMG-1), lipopolysaccharide (LPS), or HMGB 1 B box (B box).
  • HMG-1 HMGBl
  • LPS lipopolysaccharide
  • B box HMGB 1 B box
  • FIG. 11B is a histogram ofthe effect of HMGBl (HMG-1) or HMGBl B box (B Box) polypeptide stimulation on activation ofthe NF- ⁇ B-dependent ELAM promoter (measured by luciferase activity) in RAW 264.7 cells co-transfected with a murine MyD 88 -dominant negative (+MyD 88 DN) mutant (corresponding to amino acids 146-296), or empty vector (-MyD 88 DN). Data are expressed as the ratio
  • FIG. 12A is the amino acid sequence of a human HMG1 polypeptide (SEQ IDNO:l).
  • FIG. 12B is the amino acid sequence of rat and mouse HMG1 (SEQ ID NO:l).
  • FIG. 12C is the amino acid sequence of human HMG2 (SEQ ID NO:3).
  • FIG. 12D is the amino acid sequence of a human, mouse, and rat HMG1 A box polypeptide (SEQ ID NO:4).
  • FIG. 12E is the amino acid sequence of a human, mouse, and rat HMG1 B box polypeptide (SEQ ID NO:5).
  • FIG. 12F is the nucleic acid sequence of a forward primer for human HMG1 (SEQ ID NO:6).
  • FIG. 12G is the nucleic acid sequence of a reverse primer for human HMG1 (SEQ ID NO:7).
  • FIG. 12H is the nucleic acid sequence of a forward primer for the carboxy terminus mutant of human HMG1 (SEQ ID NO:8).
  • FIG. 121 is the nucleic acid sequence of a reverse primer for the carboxy terminus mutant of human HMG1 (SEQ ID NO:9).
  • FIG. 12J is the nucleic acid sequence of a forward primer for the amino terminus plus B box mutant of human HMG1 (SEQ ID NO:10).
  • FIG. 12K is the nucleic acid sequence of a reverse primer for the amino terminus plus B box mutant of human HMG1 (SEQ ID NO: 11).
  • FIG. 12L is the nucleic acid sequence of a forward primer for a B box mutant of human HMG1 (SEQ ID NO: 12).
  • FIG. 12M is the nucleic acid sequence of a reverse primer for a B box mutant of human HMG1 (SEQ ID NO: 13).
  • FIG. 12N is the nucleic acid sequence of a forward primer for the amino terminus plus A box mutant of human HMG1 (SEQ ID NO: 14).
  • FIG. 120 is the nucleic acid sequence of a reverse primer for the amino terminus plus A box mutant of human HMG1 (SEQ ID NO:15).
  • FIG. 13 is a sequence alignment of HMGBl polypeptide sequences from rat (SEQ ID NO:2), mouse (SEQ ID NO:2), and human (SEQ ID NO: 18).
  • FIG. 14A is the nucleic acid sequence of HMG1L5 (formerly HMG1L10) (SEQ ID NO: 32) encoding an HMGB polypeptide.
  • FIG. 14B is the polypeptide sequence of HMG1L5 (formerly HMG1L10)
  • FIG. 14C is the nucleic acid sequence of HMG1L1 (SEQ ID NO: 33) encoding an HMGB polypeptide.
  • FIG. 14D is the polypeptide sequence of HMG1L1 (SEQ ID NO: 25) encoding an HMGB polypeptide.
  • FIG. 14E is the nucleic acid sequence of HMG1L4 (SEQ ID NO: 34) encoding an HMGB polypeptide.
  • FIG. 14F is the polypeptide sequence of HMG1L4 (SEQ ID NO: 26) encoding an HMGB polypeptide.
  • FIG. 14G is the nucleic acid sequence ofthe HMG polypeptide sequence of the BAC clone RPl 1-395A23 (SEQ ID NO: 35).
  • FIG. 14H is the polypeptide sequence ofthe HMG polypeptide sequence of the BAC clone RPl 1-395A23 (SEQ ID NO: 27) encoding an HMGB polypeptide.
  • FIG. 141 is the nucleic acid sequence of HMG1L9 (SEQ ID NO: 36) encoding an HMGB polypeptide.
  • FIG. 14J is the polypeptide sequence of HMG1L9 (SEQ ID NO: 28) encoding an HMGB polypeptide.
  • FIG. 14K is the nucleic acid sequence of LOC122441 (SEQ ID NO: 37) encoding an HMGB polypeptide.
  • FIG. 14L is the polypeptide sequence of LOC122441 (SEQ ID NO: 29) encoding an HMGB polypeptide.
  • FIG. 14M is the nucleic acid sequence of LOC139603 (SEQ ID NO: 38) encoding an HMGB polypeptide.
  • FIG. 14N is the polypeptide sequence of LOC139603 (SEQ ID NO: 30) encoding an HMGB polypeptide.
  • FIG. 14O is the nucleic acid sequence of HMG1L8 (SEQ ID NO: 39) encoding an HMGB polypeptide.
  • FIG. 14P is the polypeptide sequence of HMG1L8 (SEQ ID NO: 31) encoding an HMGB polypeptide.
  • the present invention is based on a series of discoveries that further elucidate various characteristics ofthe ability of HMGBl to induce production of proinflammatory cytokines and inflammatory cytokine cascades.
  • the proinflammatory active domain of HMGBl is the B box (and in particular, the first 20 amino acids ofthe B box), and that antibodies specific to the B box will inhibit proinflammatory cytokine release and inflammatory cytokine cascades, with results that can alleviate deleterious symptoms caused by inflammatory cytokine cascades.
  • the A box is a weak agonist of inflammatory cytokine release, and competitively inhibits the proinflammatory activity ofthe B box and of HMGBl .
  • inhibitors of TNF biological activity can be combined with HMGB A boxes and/or antibodies to HMGB 1 , to form pharmaceutical compositions for use in treating conditions characterized by activation of an inflammatory cytokine cascade in patients.
  • an "HMGB polypeptide” or an “HMGB protein” is a substantially pure, or substantially pure and isolated polypeptide, that has been separated from components that naturally accompany it, or a synthetically or recombinantly produced polypeptide having the same amino acid sequence, and increases inflammation, and/or increases release of a proinflammatory cytokine from a cell, and/or increases the activity ofthe inflammatory cytokine cascade.
  • the HMGB polypeptide has one ofthe above biological activities.
  • the HMGB polypeptide has two ofthe above biological activities.
  • the HMGB polypeptide has all three ofthe above biological activities.
  • the HMGB polypeptide is a mammalian HMGB polypeptide, for example, a human HMGBl polypeptide.
  • an HMGB polypeptide include a polypeptide comprising or consisting ofthe sequence of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, or SEQ ID NO: 18.
  • the HMGB polypeptide contains a B box DNA binding domain and/or an A box DNA binding domain, and/or an acidic carboxyl tenninus as described herein.
  • HMGB polypeptides are described in GenBank Accession Numbers AAA64970, AAB08987, P07155, AAA20508, S29857, P09429, NP_002119, CAA31110, S02826, U00431, X67668, NP_005333, NM_016957, and J04179, the entire teachings of which are incorporated herein by reference.
  • HMGB polypeptides include, but are not limited to mammalian HMG1 ((HMGBl) as described, for example, in GenBank Accession Number U51677), HMG2 ((HMGB 2) as described, for example, in GenBanlc Accession Number M83665), HMG-2A ((HMGB3, HMG-4) as described, for example, in GenBank Accession Numbers NM_005342 andNP_005333), HMG14 (as described, for example, in GenBank Accession Number P05114), HMG17 (as described, for example, in GenBank Accession Number X13546), HMGI (as described, for example, in GenBanlc Accession Number L17131), and HMGY (as described, for example, in GenBank Accession Number M23618); nonmammalian HMG Tl (as described, for example, in GenBank Accession Number X02666) and HMG T2 (as described, for example, in GenBank Accession Number L328
  • HMGB proteins are polypeptides encoded by HMGB nucleic acid sequences having GenBank Accession Numbers NG_000897 (HMG1L5 (formerly HMG1L10)) (and in particular by nucleotides 150-797 of NG_000897, as shown in FIGS. 14A and 14B); AF076674 (HMG1L1) (and in particular by nucleotides 1-633 of AF076674, as shown in FIGS. 14C and 14D; AF076676 (HMG1L4) (and in particular by nucleotides 1-564 of AF076676, as shown in FIGS.
  • AC010149 HMG sequence from BAC clone RPl 1-395A23 (and in particular by nucleotides 75503-76117 of AC010149), as shown in FIGS. 14G and 14H); AF165168 (HMG1L9) (and in particular by nucleotides 729-968 of AF165168, as shown in FIGS. 141 and 14J); XM_063129 (LOC122441) (and in particular by nucleotides 319-558 of XM_063129, as shown in FIGS.
  • XM_066789 LOC139603
  • AF165167 HMG1L8
  • HMGB polypeptides ofthe present invention also encompass sequence variants.
  • Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other variants.
  • Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by an HMGB nucleic acid molecule, and complements and portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising the nucleotide sequence of an HMGB nucleic acid molecule.
  • Examples of HMGB nucleic acid molecules are known in the art and can be derived from HMGB polypeptides as described herein.
  • Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods.
  • the HMGB polypeptide has at least 60%, more preferably, at least 70%, 75%, 80%, 85%, or 90%, and most preferably at least 95%, sequence identity to a sequence selected from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3 and SEQ ID NO.T8, as determined using the BLAST program and parameters described herein and one of more of the biological activities of an HMGB polypeptide.
  • the present invention is directed to an HMGB polypeptide fragment that has HMGB biological activity.
  • an HMGB polypeptide fragment that has HMGB biological activity or a “biologically active HMGB fragment” is meant a fragment of an HMGB polypeptide that has the activity of an HMGB polypeptide.
  • An example of such an HMGB polypeptide fragment is the HMGB B box, as described herein.
  • Biologically active HMGB fragments can be generated using standard molecular biology techniques and assaying the function of the fragment by determining if the fragment, when administered to a cell, increases release of a proinflammatory cytokine from the cell, compared to a suitable control, for example, using methods described herein.
  • an "HMGB A box”, also referred to herein as an “A box”, is a substantially pure, or substantially pure and isolated polypeptide, that has been separated from components that naturally accompany it, and consists of an amino acid sequence that is less than a full length HMGB polypeptide and which has one or more ofthe following biological activities: inhibiting inflammation, and/or inhibiting release of a proinflammatory cytokine from a cell, and/or decreasing the activity of the inflammatory cytokine cascade.
  • the HMGB A box polypeptide has one ofthe above biological activities.
  • the HMGB A box polypeptide has two ofthe above biological activities.
  • the HMGB A box polypeptide has all three ofthe above biological activities.
  • the HMGB A box has no more than 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, ofthe biological activity of a full length HMGB polypeptide.
  • the HMGB A box amino acid consists ofthe sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal.
  • An HMGB A box is also a recombinantly produced polypeptide having the same amino acid sequence as the A box sequences described above.
  • the HMGB A box is a mammalian HMGB A box, for example, a human HMG1 A box.
  • the HMGB A box polypeptides ofthe present invention preferably comprise or consist ofthe sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal.
  • An HMGB A box often has no more than about 85 amino acids and no fewer than about 4 amino acids.
  • polypeptides having A box sequences within them include, but are not limited to, the HMGB proteins and polypeptides described herein.
  • the A box sequences in such polypeptides can be determined and isolated using methods described herein, for example, by sequence comparisons to A boxes described herein and testing for A box biological activity using methods described herein or other methods known in the art.
  • HMGB A box polypeptide sequences include the following sequences: PDASVNFSEF SKKCSERWKT MSAKEKGKFE
  • DMAKADKARY EREMKTYIPP KGET human HMGBl; SEQ ID NO: 40
  • DSSVNFAEF SKKCSERWKT MSAKEKSKFE DMAKSDKARY DREMKNYVPP KGDK human HMGB2; SEQ ID NO: 41
  • PEVPVNFAEF SKKCSERWKT VSGKEKSKFD EMAKADKVRY DREMKDYGPA KGGK human HMGB3; SEQ ID NO: 42
  • PDASVNFSEF SKKCSERWKT MSAKEKGKFE DMAKADKARY EREMKTYIPP KGET HMG1L5 (formerly HMG1L10); SEQ ID NO: 43
  • SDASVNFSEF SNKCSERWKT MSAKEKGKFE DMAKADKTHY ERQMKTYIPP KGET HMG1L1; SEQ ID NO: 44
  • PDASVNFSEF SKKCSERWKA MSAKDKGKFE DMAKVDKADY EREMKTYIPP KGET
  • the HMGB A box polypeptides ofthe present invention also encompass sequence variants.
  • Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other variants.
  • Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by an HMGB A box nucleic acid molecule, and complements and portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising the nucleotide sequence of an HMGB A box nucleic acid molecule.
  • HMGB A box nucleic acid molecules are known in the art and can be derived from HMGB A polypeptides as described herein. Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods.
  • an HMGB A box has at least 60%, more preferably, at least 70%, 75%, 80%, 85%, or 90%, and most preferably at least 95%, sequence identity to an HMGB A box polypeptide described herein, for example, the sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57, as determined using the BLAST program and parameters described herein, and one of more ofthe biological activities of an HMGB A box, as determined using methods described herein or other method known in the art.
  • the present invention also features A box biologically active fragments.
  • an "A box fragment that has A box biological activity” or an "A box biologically active fragment” is meant a fragment of an HMGB A box that has the activity of an HMGB A box, as described herein.
  • the A box fragment can decrease release of a pro-inflammatory cytokine from a vertebrate cell, decrease inflammation, and/or decrease activity ofthe inflammatory cytokine cascade.
  • a box fragments can be generated using standard molecular biology techniques and assaying the function ofthe fragment by determining if the fragment, when administered to a cell inhibits release of a proinflammatory cytokine from the cell, for example, using methods described herein.
  • a box biologically active fragments can be used in the methods described herein in which full length A box polypeptides are used, for example, inhibiting release of a proinflammatory cytokine from a cell, or treating a patient having a condition characterized by activation of an inflammatory cytokine cascade.
  • an "HMGB B box”, also referred to herein as a "B box” is a substantially pure, or substantially pure and isolated polypeptide, that has been separated from components that naturally accompany it, and consists of an amino acid sequence that is less than a full length HMGB polypeptide and has one or more ofthe following biological activities: increasing inflammation, increasing release of a proinflammatory cytokine from a cell, and or increasing the activity ofthe inflammatory cytokine cascade.
  • the HMGB B box polypeptide has one ofthe above biological activities.
  • the HMGB B box polypeptide has two ofthe above biological activities.
  • the HMGB B box polypeptide has all three ofthe above biological activities.
  • the HMGB B box has at least 25%, 30%, 40%, 50%, 60%, 70%, 80% or 90%, ofthe biological activity of a full length HMGB polypeptide.
  • the HMGB B box does not comprise an HMGB A box.
  • the HMGB B box is a polypeptide that is about 90%, 80%, 70%, 60%, 50%, 40%, 35%, 30%, 25%, or 20%, ofthe length of a full length HMGB 1 polypeptide.
  • the HMGB B box comprises or consists ofthe sequence of SEQ ID NO:5, SEQ ID NO:20 or SEQ ID NO:58, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal, but is still less than the full length HMGB polypeptide.
  • An HMGB B box polypeptide is also a recombinantly produced polypeptide having the same amino acid sequence as an HMGB B box polypeptide described above.
  • the HMGB B box is a mammalian HMGB B box, for example, a human HMGBl B box.
  • An HMGB B box often has no more than about 85 amino acids and no fewer than about 4 amino acids.
  • polypeptides having B box sequences within them include, but are not limited to, the HMGB proteins and polypeptides described herein.
  • the B box sequences in such polypeptides can be determined and isolated using methods described herein, for example, by sequence comparisons to B boxes described herein and testing for biological activity, using methods described herein or other methods known in the art.
  • HMGB B box polypeptide sequences include the following sequences: FKDPNAPKRP PSAFFLFCSE YRPKIKGEHP GLSIGDNAKK LGEMW ⁇ TAA DDKQPYEKKA AKLKEKYEKD IAAY (human HMGB 1 ; SEQ ID NO: 51); KKDPNAPKRP PSAFFLFCSE HRPKIKSEHP GLSIGDTAKK LGEMWSEQSA KDKQPYEQKA AKLKEKYEKD IAAY (human HMGB2; SEQ ID NO: 52); FKDPNAPKRL PSAFFLFCSE YRPKIKGEHP GLSIGDNAKK LGEMW ⁇ TAA DDKQPYEKKA AKLKEKYEKD IAAY (HMG1L5 (formerly HMG1L10); SEQ ID NO: 53); FKDPNAPKRP PSAFFLFCSE YHPKIKGEHP GLSIGDNAKK LGEMW ⁇ TAA DDKQPGEKKA AKL
  • HMGB B box polypeptides ofthe invention also encompass sequence variants.
  • Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other variants.
  • Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by an HMGB box nucleic acid molecule, and complements and portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising the nucleotide sequence of an HMGB B box nucleic acid molecule.
  • HMGB B box nucleic acid molecules are known in the art and can be derived from HMGB B box polypeptides as described herein.
  • Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog.
  • Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis.
  • Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods.
  • a non-naturally occurring HMGB B box polypeptide has at least 60%, more preferably, at least 70%, 75%, 80%, 85%, or 90%, and most preferably at least 95%, sequence identity to the sequence of an HMGB B box as described herein, for example, the sequence of SEQ ID NO:5, SEQ ID NO:20, or SEQ ID NO:58, as determined using the BLAST program and parameters described herein.
  • the HMGB B box consists ofthe sequence of SEQ ID NO:5, SEQ ID NO:20, or SEQ ID NO:58, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal, and has one or more ofthe biological activities of an HMGB B box, as detennined using methods described herein or other methods known in the art.
  • the present invention is directed to a polypeptide comprising an HMGB B box biologically active fragment that has B box biological activity, or a non-naturally occurring HMGB B box fragment
  • the present invention is directed to a polypeptide comprising a vertebrate HMGB B box or a fragment thereof that has B box biological activity, or a non-naturally occurring HMGB B box but not comprising a full length HMGB polypeptide.
  • a "B box fragment that has B box biological activity” or a "B box biologically active fragment” is meant a fragment of an HMGB B box that has the activity of an HMGB B box.
  • the B box fragment can induce release of a pro-inflammatory cytokine from a vertebrate cell or increase inflammation, or induce the inflammatory cytokine cascade.
  • a B box fragment is the fragment comprising the first 20 amino acids ofthe HMGBl B box (SEQ ID NO: 16 or SEQ ID NO:23), as described herein.
  • B box fragments can be generated using standard molecular biology techniques and assaying the function ofthe fragment by determining if the fragment, when administered to a cell, increases release of a proinflammatory cytokine from the cell, as compared to a suitable control, for example, using methods described herein or other methods known in the art.
  • HMGB polypeptides, HMGB A boxes, and HMGB B boxes include polypeptides that have sequence identity to the HMGB polypeptides, HMGB A boxes, and HMGB B boxes described herein.
  • two polypeptides are substantially homologous or identical when the amino acid sequences are at least about 60%, 70%, 75%, 80%, 85%, 90%, or 95% or more, homologous or identical.
  • the percent identity of two amino acid sequences (or two nucleic acid sequences) can be determined by aligning the sequences for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first sequence).
  • the length ofthe HMGB polypeptide, HMGB A box polypeptide, or HMGB B box polypeptide aligned for comparison purposes is at least 30%), preferably, at least 40%, more preferably, at least 60%, and even more preferably, at least 70%>, 80%>, 90%, or 100%, ofthe length ofthe reference sequence, for example, those sequence provided in FIGS. 12A-12E, FIGS. 14A-14P, and SEQ ID NOS: 18, 20, and 22.
  • the database searched is a non-redundant (NR) database
  • parameters for sequence comparison can be set at: no filters; Expect value of 10; Word Size of 3; the Matrix is BLOSUM62; and Gap Costs have an Existence of 11 and an Extension of 1.
  • the percent identity between two amino acid sequences can be accomplished using the GAP program in the GCG software package (Accelrys, San Diego, California) using either a Blossom 63 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4.
  • the percent identity between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package (Accelrys, San Diego, California), using a gap weight of 50 and a length weight of 3.
  • cytokine is a soluble protein or peptide which is naturally produced by mammalian cells and which acts in vivo as a humoral regulator at micro- to picomolar concentrations. Cytokines can, either under normal or pathological conditions, modulate the functional activities of individual cells and tissues.
  • a proinflammatory cytokine is a cytokine that is capable of causing any ofthe following physiological reactions associated with inflammation: vasodilation, hyperemia, increased permeability of vessels with associated edema, accumulation of granulocytes and mononuclear phagocytes, or deposition of fibrin.
  • the proinflammatory cytokine can also cause apoptosis, such as in chronic heart failure, where TNF has been shown to stimulate cardiomyocyte apoptosis (Pulkki, Ann. Med. 29: 339-343, 1997; and Tsutsui et al, Immunol. Rev. 174:192-209, 2000).
  • proinflammatory cytokines are tumor necrosis factor (TNF), interleulcin (IL)-l , IL-l ⁇ , IL-6, IL-8, IL-18, interferon ⁇ , HMG-1, platelet-activating factor (PAF), and macrophage migration inhibitory factor (MIF).
  • Proinflammatory cytokines are to be distinguished from anti-inflammatory cytokines, such as IL-4, IL-10, and IL-13, which are not mediators of inflammation.
  • proinflammatory cytokines are produced in an inflammatory cytokine cascade, defined herein as an in vivo release of at least one proinflammatory cytokine in a mammal, wherein the cytokine release affects a physiological condition ofthe mammal.
  • an inflammatory cytokine cascade is inhibited in embodiments of the invention where proinflammatory cytokine release causes a deleterious physiological condition.
  • an agent that inhibits TNF biological activity is an agent that decreases one or more ofthe biological activities of TNF.
  • TNF biological activity include, but are not limited to, vasodilation, hyperemia, increased permeability of vessels with associated edema, accumulation of granulocytes and mononuclear phagocytes, and deposition of fibrin.
  • Agents that inhibit TNF biological activity include agents that inhibit (decrease) the interaction between TNF and a TNF receptor. Examples of such agents include antibodies or antigen binding fragments thereof that bind to TNF, antibodies or antigen binding fragments that bind a TNF receptor, and molecules that bind TNF or the TNF receptor and prevent
  • agents include, but are not limited to peptides, proteins, synthesized molecules, for example, synthetic organic molecules, naturally-occurring molecule, for example, naturally occurring organic molecules, nucleic acid molecules, and components thereof.
  • agents that inhibit TNF biological activity include infliximab (Remicade; Centocor, Inc.,
  • Inflammatory cytokine cascades contribute to deleterious characteristics, including inflammation and apoptosis, of numerous disorders. Included are disorders characterized by both localized and systemic reactions, including, without limitation, the disorders described herein (e.g., those conditions enumerated in the background section of this specification).
  • Particular disorders characterized by inflammatory cytokine cascades include, e.g., sepsis, allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstructive pulmonary disease, psoriasis, pancreatitis, peritonitis, burns, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative colitis, multiple sclerosis, and cachexia.
  • sepsis allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstructive pulmonary disease, psoriasis, pancreatitis, peritonitis, burns, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative co
  • the present invention is directed to a polypeptide composition comprising a vertebrate HMGB A box, or a biologically active fragment thereof, which can inhibit release of a piOinflammatory cytokine from a cell treated with HMG, or which can be used to treat a condition characterized by activation of an inflammatory cytokine cascade.
  • the invention is directed to compositions comprising an HMGB A box, or a biologically active fragment or variant thereof, in combination with one or more agents that inhibit TNF biological activity, for example, infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, or Thalidomide.
  • agents that inhibit TNF biological activity for example, infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, or Thalidomide.
  • Such compositions can be used to inhibit release of a proinflammatory cytokine from a vertebrate cell treated with HMG, and/or can be used to treat a condition characterized by activation of an inflammatory cytokine cascade.
  • the use ofthe terms “inhibit” or “decrease” encompasses at least a small but measurable reduction in proinflammatory cytokine release.
  • the release ofthe proinflammatory cytokine is inhibited by at least 20% over non-treated controls; in more preferred embodiments, the inhibition is at least 50%; in still more preferred embodiments, the inhibition is at least 70%, and in the most preferred embodiments, the inhibition is at least 80%. Inhibition can be assessed using methods described herein or other methods known in the art. Such reductions in proinflammatory cytokine release are capable of reducing the deleterious effects of an inflammatory cytokine cascade in in vivo embodiments.
  • HMGB A boxes e.g., vertebrate HMGB A boxes
  • an HMGB A box e.g., a vertebrate HMGB A box
  • an HMGB A box is within the scope ofthe invention.
  • the HMGB A box is a vertebrate HMGB A box (e.g., a mammalian HMGB A box, such as a human HMGB 1 A box provided herein as SEQ ID NO :4, SEQ ID NO:22, or SEQ ID NO:57). Also included in the present invention are fragments ofthe HMGBl A box having HMGB A box biological activity, as described herein.
  • a vertebrate HMGB A box e.g., a mammalian HMGB A box, such as a human HMGB 1 A box provided herein as SEQ ID NO :4, SEQ ID NO:22, or SEQ ID NO:57.
  • fragments ofthe HMGBl A box having HMGB A box biological activity as described herein.
  • non-naturally occurring HMGB A boxes can be created without undue experimentation, which would inhibit release of a proinflammatory cytokine from a vertebrate cell treated with a vertebrate HMGB.
  • These non-naturally occurring functional A boxes can be created by aligning amino acid sequences of HMGB A boxes from different sources, and making one or more substitutions in one ofthe sequences at amino acid positions where the A boxes differ. The substitutions are preferably made using the same amino acid residue that occurs in the compared A box. Alternatively, a conservative substitution is made from either ofthe residues.
  • Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains.
  • Conservatively substituted amino acids can be grouped according to the chemical properties of their side chains. For example, one grouping of amino acids includes those amino acids have neutral and hydrophobic side chains (a, v, 1, i, p, w, f, and m); another grouping is those amino acids having neutral and polar side chains (g, s, t, y, c, n, and q); another grouping is those amino acids having basic side chains (k, r, and h); another grouping is those amino acids having acidic side chains (d and e); another grouping is those amino acids having aliphatic side chains (g, a, v, 1, and i); another grouping is those amino acids having aliphatic-hydroxyl side chains (s and t); another grouping is those amino acids having amine-containing side chains (n, q, k, r, and h); another grouping is those amino acids having aromatic side chains (f,
  • HMGB A box polypeptide While a conservative amino acid substitution would be expected to preserve the biological activity of an HMGB A box polypeptide, the following is one example of how non-naturally occurring A box polypeptides (variants) can be made by comparing the human HMGB 1 A box (SEQ ID NO :4) with residues 32 to 85 of SEQ ID NO:3 ofthe human HMGB2 A box (SEQ ID NO:17).
  • a non-naturally occurring HMGB A box can be created by, for example, by substituting the alanine (a) residue at the third position in the HMGBl A box with the serine (s) residue that occurs at the third position ofthe HMGB2 A box.
  • the skilled artisan would know that the substitution would provide a functional non- naturally occurring A box because the s residue functions at that position in the HMGB2 A box.
  • the third position ofthe HMGBl A box can be substituted with any amino acid that is conservative to alanine or serine, such as glycine (g), threonine (t), valine (v) or leucine (1).
  • g glycine
  • t threonine
  • v valine
  • leucine leucine
  • HMGB A boxes could be created without undue experimentation wliich would be expected to be functional, and the functionality of any particular non-naturally occurring HMGB A box could be predicted with adequate accuracy.
  • the functionality of any non-naturally occurring HMGB A box could be determined without undue experimentation by simply adding it to cells along with an HMGB polypeptide, and determining whether the A box inhibits release of a proinflammatory cytokine by the cells, using, for example, methods described herein.
  • the cell from which the A box or an A box biologically active fragment will inhibit the release of HMG-induced proinflammatory cytokines can be any cell that can be induced to produce a proinflammatory cytokine.
  • the cell is a mammalian cell, for example, an immune cell (e.g., a macrophage, a monocyte, or a neutrophil).
  • Polypeptides comprising an A box or A box biologically active fragment that can inhibit the production of any single proinflammatory cytokine, now known or later discovered, are within the scope ofthe present invention.
  • the antibodies can inhibit the production of TNF, IL- 1 ⁇ , and/or IL-6.
  • the antibodies can inhibit the production of any proinflammatory cytokines produced by the vertebrate cell.
  • the present invention is directed to a polypeptide composition
  • a polypeptide composition comprising a vertebrate HMGB B box, or a biologically active fragment thereof, which can increase release of a proinflammatory cytokine from a vertebrate cell treated with HMGB.
  • the use ofthe term "increase” encompasses at least a small but measurable rise in proinflammatory cytokine release.
  • the release ofthe proinflammatory cytokine is increased by at least 1.5-fold, at least 2-fold, at least 5-fold, or at least 10-fold, over non-treated controls.
  • Such increases in proinflammatory cytokine release are capable of increasing the effects of an inflammatory cytokine cascade in in vivo embodiments.
  • Such polypeptides can also be used to induce weight loss and/or treat obesity.
  • the B box comprises SEQ ID NO:5, SEQ ID NO: 20 or SEQ ID NO:58, which are the sequences (three different lengths) ofthe human HMGBl B box, or, comprises the B box sequences from the polypeptides shown in FIGS.
  • HMGB B box 14A-14P is a fragment of an HMGB B box that has B box biological activity.
  • a 20 amino acid sequence contained within SEQ ID NO: 20 contributes to the function ofthe B box.
  • This 20 amino acid B-box fragment has the following amino acid sequence: flcdpnapkrl psafflfcse (SEQ ID NO:23).
  • Another example of an HMGB B box biologically active fragment consists of amino acids 1-20 of SEQ ID NO: 5 (naplcrppsafflfcseyrpk; SEQ ID NO: 16).
  • Antibodies to HMGB and HMGB B Box Polypeptides The invention is also directed to a purified preparation of antibodies that bind to an HMGB polypeptide or a biologically active fragment thereof (anti-HMGB antibodies).
  • the anti-HMGB antibodies can be neutralizing antibodies (i.e., can inhibit a biological activity of an HMG polypeptide or a biologically active fragment thereof, for example, the release of a proinflammatory cytokine from a vertebrate cell induced by HMG).
  • the invention is also directed to a purified preparation of antibodies that specifically bind to a vertebrate high mobility group protein (HMG) B box or a biologically active fragment thereof, but do not selectively bind to non-B box epitopes of HMGB (anti-HMGB B box antibodies).
  • HMG high mobility group protein
  • the antibodies can also be neutralizing antibodies (i.e., they can inhibit a biological activity of a B box polypeptide or biologically active fragment thereof, for example, the release of a proinflammatory cytokine from a vertebrate cell induced by HMGB).
  • Such antibodies can be combined with one or more agents that inhibit TNF biological activity, for example, infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, or Thalidomide.
  • antibody or “purified antibody” as used herein refers to immunoglobulm molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that selectively binds an antigen.
  • a molecule that selectively binds to a polypeptide ofthe invention is a molecule that binds to that polypeptide or a fragment thereof, but does not substantially bind other molecules in a sample, e.g., a biological sample that naturally contains the polypeptide.
  • the antibody is at least 60%, by weight, free from proteins and naturally occurring organic molecules with which it is naturally associated. More preferably, the antibody preparation is at least 75% or 90%, and most preferably, 99%, by weight, antibody.
  • immunologically active portions of immunoglobulin molecules include F(ab) and F(ab') 2 fragments that can be generated by treating the antibody with an enzyme such as pepsin.
  • the invention provides polyclonal and monoclonal antibodies that selectively bind to a HMGB B box polypeptide ofthe invention.
  • a monoclonal antibody composition thus typically displays a single binding affinity for a particular polypeptide ofthe invention with which it immunoreacts.
  • Polyclonal antibodies can be prepared, e.g., as described herein, by immunizing a suitable subject with a desired immunogen, e.g., an HMGB B box polypeptide ofthe invention or fragment thereof.
  • the antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide.
  • ELISA enzyme linked immunosorbent assay
  • the antibody molecules directed against the polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (Nature 256:495-497, 1975), the human B cell hybridoma technique (Kozbor et al., Immunol. Today 4:72, 1983), the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96, 1985) or trioma techniques.
  • standard techniques such as the hybridoma technique originally described by Kohler and Milstein (Nature 256:495-497, 1975), the human B cell hybridoma technique (Kozbor et al., Immunol. Today 4:72, 1983), the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96, 1985
  • hybridomas The technology for producing hybridomas is well known (see generally Current Protocols in Immunology, Coligan et al, (eds.) John Wiley & Sons, Inc., New York, NY, 1994). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an immunogen as described above, and the culture supernatants ofthe resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds a particular polypeptide (e.g., a polypeptide ofthe invention).
  • lymphocytes typically splenocytes
  • a monoclonal antibody to an HMGB B box polypeptide ofthe invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide to thereby isolate immunoglobulin library members that bind the polypeptide.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAPTM Phage Display Kit, Catalog No. 240612).
  • recombinant antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope ofthe invention.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art.
  • antibodies ofthe invention e.g., a monoclonal antibody
  • a polypeptide-specific antibody can facilitate the purification of natural polypeptide from cells and of recombinantly produced polypeptide expressed in host cells.
  • an antibody specific for an HMGB B box polypeptide ofthe invention can be used to detect the polypeptide (e.g., in a cellular lysate, cell supernatant, or tissue sample) in order to evaluate the abundance and pattern of expression ofthe polypeptide.
  • vertebrate HMGB polypeptides and HMGB B boxes show a high degree of sequence conservation, it is believed that vertebrate HMGB polypeptides or HMGB B boxes in general can induce release of a proinflammatory cytokine from a vertebrate cell. Therefore, antibodies against vertebrate HMGB polypeptides or HMGB B boxes are within the scope ofthe invention. In one embodiment, the antibodies are neutralizing antibodies.
  • the HMGB polypeptide is a mammalian HMG, as described herein, more preferably a mammalian HMGBl polypeptide, most preferably a human HMGB 1 polypeptide, provided herein as SEQ ID NO: 1.
  • Antibodies can also be directed against an HMGB polypeptide fragment that has HMGB polypeptide biological activity.
  • the HMGB B box is a mammalian HMGB B box, more preferably a mammalian HMGBl B box, most preferably a human HMGBl B box, provided herein as SEQ ID NO:5, SEQ ID NO:20, or SEQ ID NO:58.
  • Antibodies can also be directed against an HMGB B box fragment that has B box biological activity.
  • Antibodies generated against an HMGB immunogen or an HMGB B box immunogen can be obtained by administering an HMGB polypeptide, or fragment thereof, an HMGB B box or fragment thereof, or cells comprising the HMGB polypeptide, the HMGB B box, or fragments thereof, to an animal, preferably a nonhuman, using routine protocols.
  • the polypeptide such as an antigenically or immunologically equivalent derivative, is used as an antigen to immunize a mouse or other animal, such as a rat or chicken.
  • the immunogen may be associated, for example, by conjugation, with an immunogenic carrier protein, for example, bovine serum albumin (BSA) or keyhole limpet haemocyanin (KLH).
  • BSA bovine serum albumin
  • KLH keyhole limpet haemocyanin
  • a multiple antigenic peptide comprising multiple copies ofthe HMGB or HMGB B box or fragment, may be sufficiently antigenic to improve immunogenicity so as to obviate the need for a carrier.
  • Bispecific antibodies having two antigen binding domains where each is directed against a different HMGB or HMGB B box epitope, may also be produced by routine methods.
  • any technique known in the art that provides antibodies produced by continuous cell line cultures can be used. See, e.g., Kohler and Milstein, supra; and Cole et al, supra.
  • the antibody is preferably modified to make it less immunogenic in the individual.
  • the individual is human the antibody is preferably "humanized"; where the complementarity determining region(s) ofthe antibody is transplanted into a human antibody (for example, as described in Jones et al. (Nature 321:522-525, 1986); and Tempest et al. (Biotechnology 9:266-273, 1991)).
  • Phage display technology can also be utilized to select antibody genes with binding activities towards the polypeptide either from repertoires of PCR amplified v-genes of lymphocytes from humans screened for possessing anti-B box antibodies or from naive libraries (McCafferty et al., Nature 348:552-554, 1990; and Marks, et al., Biotechnology 10:779-783, 1992).
  • the affinity of these antibodies can also be improved by chain shuffling (Clackson et al., Nature 352: 624-628, 1991).
  • Anti-HMGB B box antibodies that can inhibit the production of any single proinflammatory cytokine, and/or inhibit the release of a proinflammatory cytokine from a cell, and/or inhibit a condition characterized by activation of an inflammatory cytokine cascade, are within the scope ofthe present invention.
  • the antibodies can inhibit the production of TNF, IL-1 ⁇ , and/or IL-6.
  • the antibodies can inhibit the production of any proinflammatory cytokines produced by the vertebrate cell.
  • the cell can be any cell that can be induced to produce a proinflammatory cytokine.
  • the cell is an immune cell, for example, macrophages, monocytes, or neutrophils.
  • Compositions Comprising One or More of an HMGB A box polypeptide, an Antibody to HMGB, an Antibody to an HMGB B box, and an Inhibitor of TNF Biological Activity
  • the present invention is directed to a composition
  • a composition comprising any ofthe above-described polypeptides (e.g., an HMGB A box polypeptide or biologically active fragment as described herein) in a pharmaceutically acceptable carrier.
  • suitable pharmaceutically acceptable carriers include those described herein.
  • the composition can inhibit a condition characterized by activation of an inflammatory cytokine cascade.
  • the condition can be one where the inflammatory cytokine cascade causes a systemic reaction, such as with endotoxic shock.
  • the condition can be mediated by a localized inflammatory cytokine cascade, as in rheumatoid arthritis.
  • Nonlimiting examples of conditions which can be usefully treated using the present invention include those conditions enumerated in the background section of this specification.
  • the condition to be treated is appendicitis, peptic, gastric or duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute or ischemic colitis, hepatitis, Crohn's disease, asthma, allergy, anaphylactic shock, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, septic abortion, disseminated bacteremia, burns, Alzheimer's disease, coeliac disease, congestive heart failure, adult respiratory distress syndrome, cerebral infarction, cerebral embolism, spinal cord injury, paralysis, allograft rejection or graft- versus-host disease, hi another embodiment, the condition is endotoxic shock or allograft rejection.
  • the composition may advantageously also include an immunosuppressant that is used to inhibit allograft rejection, such as cyclosporin.
  • an immunosuppressant that is used to inhibit allograft rejection, such as cyclosporin.
  • the invention is directed to a composition comprising the antibody preparations described above (e.g., anti-HMGB B box antibodies or biologically active fragments thereof, as described herein), in a pharmaceutically acceptable carrier, hi these embodiments, the compositions can inhibit a condition characterized by the activation of an inflammatory cytokine cascade. Conditions that can be treated with these compositions have been previously enumerated.
  • the invention is directed to a composition comprising any ofthe above-described HMGB A box polypeptides, and/or an antibody or antigen binding fragment thereof that binds HMGB, and/or an antibody or antigen binding fragment thereof that binds an HMGB B box, and an agent that inhibits TNF biological activity (collectively termed "combination therapy compositions").
  • the combination therapy composition can inhibit a condition characterized by activation of an inflammatory cytokine cascade and/or inhibit release of a proinflammatory cytokine from a cell.
  • the condition can be one where the inflammatory cytokine cascade causes a systemic reaction, such as with endotoxic shock.
  • condition can be mediated by a localized inflammatory cytokine cascade, as in rheumatoid arthritis.
  • a localized inflammatory cytokine cascade as in rheumatoid arthritis.
  • Nonlimiting examples of conditions which can be usefully treated using the present invention include those conditions enumerated in the background section of this specification.
  • the condition to be treated is sepsis, allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstructive pulmonary disease, psoriasis, pancreatitis, peritonitis, burns, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative colitis, multiple sclerosis, and cachexia.
  • the combination therapy compositions are administered to a patient in need thereof in an amount sufficient to inhibit release of proinflammatory cytokine from a cell and/or to treat a condition characterized by activation of an inflammatory cytokine cascade.
  • release ofthe proinflammatory cytokine is inhibited by at least 10%, 20%, 25%, 50%, 75%, 80%, 90% or 95%, as assessed using methods described herein or other methods known in the art.
  • the carrier or excipient included with the polypeptide e.g., an HMGB A box polypeptide or biologically active fragment thereof), antibody (e.g., an anti-HMGB B box antibody or biologically active fragment thereof) or combination therapy composition (e.g., an HMGB A box polypeptide or biologically active fragment thereof and an agent that inhibits TNF biological activity, and/or an antibody or antigen binding fragment thereof that binds HMGB and an agent that inhibits TNF biological activity, and/or an antibody or antigen binding fragment thereof that binds an HMGB B box and an agent that inhibits TNF biological activity) is chosen based on the expected route of administration ofthe composition in therapeutic applications. The route of administration ofthe composition depends on the condition to be treated.
  • intravenous injection may be preferred for treatment of a systemic disorder such as endotoxic shock
  • oral administration may be preferred to treat a gastrointestinal disorder such as a gastric ulcer.
  • the route of administration and the dosage ofthe composition to be administered can be determined by the skilled artisan, without undue experimentation, in conjunction with standard dose-response studies. Relevant circumstances to be considered in making such determinations include the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response ofthe individual patient, and the severity ofthe patient's symptoms.
  • the antibody composition can be administered orally, parenterally, intranasally, vaginally, rectally, lingually, sublingually, bucally, intrabuccaly and transdermally to the patient.
  • compositions designed for oral, lingual, sublingual, buccal and intrabuccal administration can be made without undue experimentation by means well known in the art, for example, with an inert diluent or with an edible carrier.
  • the compositions may be enclosed in gelatin capsules or compressed into tablets.
  • compositions ofthe present invention may be incorporated with excipients and used in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, chewing gums and the like.
  • Tablets, pills, capsules, troches and the like may also contain binders, recipients, disintegrating agent, lubricants, sweetening agents, and flavoring agents.
  • binders include microcrystalline cellulose, gum tragacanth and gelatin.
  • excipients include starch and lactose.
  • disintegrating agents include alginic acid, corn starch and the like.
  • lubricants include magnesium stearate and potassium stearate.
  • An example of a glidant is colloidal silicon dioxide.
  • sweetening agents include sucrose, saccharin and the like.
  • flavoring agents include peppermint, methyl salicylate, orange flavoring and the like. Materials used in preparing these various compositions should be pharmaceutically pure and non-toxic in the amounts used.
  • compositions ofthe present invention can easily be administered parenterally such as, for example, by intravenous, intramuscular, intrathecal or subcutaneous injection.
  • Parenteral administration can be accomplished by incorporating the compositions ofthe present invention into a solution or suspension.
  • solutions or suspensions may also include sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol and/or other synthetic solvents.
  • Parenteral formulations may also include antibacterial agents such as, for example, benzyl alcohol and/or methyl parabens, antioxidants such as, for example, ascorbic acid and/or sodium bisulfite and chelating agents such as EDTA.
  • Buffers such as acetates, citrates and/or phosphates, and agents for the adjustment of tonicity, such as sodium chloride and/or dextrose, may also be added.
  • the parenteral preparation can be enclosed in ampules, disposable syringes or multiple dose vials made of glass or plastic.
  • Rectal administration includes administering the pharmaceutical compositions into the rectum or large intestine. This can be accomplished using suppositories or enemas.
  • Suppository formulations can easily be made by methods known in the art. For example, suppository formulations can be prepared by heating glycerin to about 120°C, dissolving the polypeptide composition, antibody composition and/or combination therapy composition in the glycerin, mixing the heated glycerin after which purified water may be added, and pouring the hot mixture into a suppository mold.
  • Transdermal administration includes percutaneous absorption ofthe composition through the skin.
  • Transdermal formulations include patches, ointments, creams, gels, salves and the like.
  • the present invention includes nasally administering to a mammal (e.g., a human) a therapeutically effective amount ofthe composition.
  • nasally administering or nasal administration includes administering the composition to the mucous membranes ofthe nasal passage or nasal cavity ofthe patient.
  • pharmaceutical compositions for nasal administration of a composition include therapeutically effective amounts ofthe polypeptide, antibody and/or combination therapy agents, prepared by well-known methods to be administered, for example, as a nasal spray, nasal drop, suspension, gel, ointment, cream or powder. Administration ofthe composition may also take place using a nasal tampon or nasal sponge.
  • compositions can also include an antagonist of an early sepsis mediator.
  • an early sepsis mediator is a proinflammatory cytokine that is released from cells soon (i.e., within 30-60 min.) after induction of an inflammatoiy cytokine cascade (e.g., exposure to LPS).
  • these cytokines are TNF, IL-l , IL-l ⁇ , IL-6, PAF, and MIF.
  • early sepsis mediators include receptors for these cytokines (for example, tumor necrosis factor receptor type 1) and enzymes required for production of these cytokines, for example, interleukin-l ⁇ converting enzyme).
  • Antagonists of any early sepsis mediator can be useful for these embodiments by further inhibiting an inflammatory cytokine cascade.
  • Nonlimiting examples of antagonists of early sepsis mediators are antisense compounds that bind to the mRNA ofthe early sepsis mediator, preventing its expression (see, e.g., Ojwang et al. (Biochemistry 36:6033-6045, 1997); Pampfer et al. (Biol. Reprod. 52:1316-1326, 1995); U.S. Patent No. 6,228,642; Yahata et al. (Antisense Nucleic Acid Drug Dev. 6:55-61, 1996); and Taylor et al. (Antisense Nucleic Acid Drug Dev.
  • ribozymes that specifically cleave the mRNA ofthe early sepsis mediator (see, e.g., Leavitt et al. (Antisense Nucleic Acid Drug Dev. 10: 409-414, 2000); Hendrix et al. (Biochem. J. 314 (Pt. 2): 655-661, 1996)), and antibodies that bind to the early sepsis mediator and inhibit their action (see, e.g., Kam and Targan (Expert Opin. Pharmacother. 1: 615-622, 2000); Nagahira et al. (J. Immunol. Methods 222, 83-92, 1999); Lavine et al. (J. Cereb.
  • agents that can be administered with the compositions described herein include, e.g., VitaxinTM and other antibodies targeting v ⁇ 3 integrin (see, e.g., U.S. Patent No. 5,753,230, PCT Publication Nos. WO 00/78815 and WO 02/070007; the entire teachings of all of which are incorporated herein by reference) and anti-IL-9 antibodies (see, e.g., PCT Publication No. WO 97/08321; the entire teachings of which are incorporated herein by reference).
  • Additional agents that can be administered with the polypeptide compositions described herein include, e.g., B7 antagonists (e.g., CTLA4Ig, anti-CD80 antibodies, anti-CD86 antibodies), methotrexate, and/or CD40 antagonists (e.g., anti-CD40 ligand (CD40L)) (see, e.g., Saito et al., J. Immunol. 160(9):4225-31 (1998)).
  • the present invention is also directed to a method of inhibiting the release of a proinflammatory cytokine from a mammalian cell.
  • the method comprises treating the cell with any ofthe HMGB A box compositions, and/or any ofthe HMGB B box or HMGB B box biologically active fragment antibody compositions, and/or any ofthe combination therapy compositions discussed above. It is believed that this method would be useful for inhibiting the cytokine release from any mammalian cell that produces a proinflammatory cytokine.
  • the cell is a macrophage, because macrophage production of proinflammatory cytokines is associated with several important diseases.
  • the proinflammatory cytokine is TNF, IL-l ⁇ , IL-1 ⁇ , MIF and/or IL-6, because those proinflammatory cytokines are particularly important mediators of disease.
  • the methods of these embodiments are useful for in vitro applications, such as in studies for determining biological characteristics of proinflammatory cytokine production in cells.
  • the preferred embodiments are in vivo therapeutic applications, where the cells are in a patient suffering from, or at risk for, a condition characterized by activation of an inflammatory cytokine cascade.
  • the present invention is directed to a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade.
  • the method comprises administering to the patient any ofthe HMGB A box compositions (including non-naturally occurring A box polypeptides and A box biologically active fragments), any ofthe HMGB B box or B box biologically active fragment antibody compositions (including non-naturally occurring B box polypeptides or biologically active fragments thereof), and/or any of the combination therapy compositions discussed above.
  • This method would be expected to be useful for any condition that is mediated by an inflammatory cytokine cascade, including any of those that have been previously enumerated.
  • preferred conditions include appendicitis, peptic, gastric or duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute or ischemic colitis, hepatitis, Crohn's disease, asthma, allergy, anaphylactic shock, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, septic abortion, disseminated bacteremia, bums, Alzheimer's disease, cerebral infarction, cerebral embolism, spinal cord injury, paralysis, allograft rejection or graft- versus-host disease.
  • the condition is endotoxic shock or allograft rejection.
  • the composition may advantageously also include an immunosuppressant that is used to inhibit allograft rejection, such as cyclosporin.
  • These methods can also usefully include the administration of an antagonist of an early sepsis mediator, an anti- ⁇ v ⁇ 3 antibody, an anti JL-9 antibody, a B7 antagonist (e.g., CTLA4Ig, an anti-CD80 antibody, an anti-CD86 antibody), methotrexate, and/or a CD40 antagonist (e.g., anti-CD40 ligand (CD40L)).
  • a B7 antagonist e.g., CTLA4Ig, an anti-CD80 antibody, an anti-CD86 antibody
  • methotrexate e.g., methotrexate
  • CD40 antagonist e.g., anti-CD40 ligand (CD40L)
  • the B box polypeptides and biologically active fragments thereof described herein can be used to induce inflammatory cytokines in the appropriate isolated cells in vitro, or ex vivo, or as a treatment in vivo.
  • the polypeptide or fragment can be administered by providing a DNA or RNA vector encoding the B box or B box fragment, with the appropriate control sequences operably linked to the encoded B box or B box fragment, so that the B box or B box fragment is synthesized in the treated cell or patient.
  • the B box polypeptides or B box fragment polypeptides or vectors as a weight loss treatment. See WO 00/47104 (the entire teachings of which are incorporated herein by reference), demonstrating that treatment with HMGBl induces weight loss.
  • the present invention is directed to methods of stimulating the release of a proinflammatory cytokine from a cell.
  • the method comprises treating the cell with any ofthe B box polypeptides or biologically active B box fragment polypeptides, for example, polypeptides that comprise or consist of the sequence of SEQ ID NO:5, SEQ ID NO:20, SEQ ID NO:58, SEQ ID NO: 16, or SEQ ID NO:23, as described herein (including non-naturally occurring B box polypeptides and fragments).
  • This method is useful for in vitro applications, for example, for studying the effect of proinflammatory cytokine production on the biology ofthe producing cell. Since the HMGB B box has the activity ofthe HMGB protein, the B box would also be expected to induce weight loss. Therefore, in additional embodiments, the present invention is a method for effecting weight loss or treating obesity in a patient.
  • the method comprises administering to the patient an effective amount of any ofthe B box polypeptides or B box fragment polypeptides described herein (including non-naturally occurring B box polypeptides and fragments).
  • the B box polypeptide or B box fragment polypeptide is in a pharmaceutically acceptable carrier.
  • the present invention is also directed to a method of determining whether a compound (test compound) inhibits inflammation and/or an inflammatory response.
  • the method comprises combining the compound with (a) a cell that releases a proinflammatory cytokine when exposed to a vertebrate HMGB B box or a biologically active fragment thereof, and (b) the HMGB B box or a biologically active fragment thereof, and then determining whether the compound inhibits the release ofthe proinflammatory cytokine from the cell, as compared to a suitable control.
  • a compound that inhibits the release ofthe proinflammatory cytokine in this assay is a compound that can be used to treat inflammation and/or an inflammatory response.
  • the HMGB B box or biologically active HMGB B box fragment can be endogenous to the cell or can be introduced into the cell using standard recombinant molecular biology techniques. Any cell that releases a proinflammatory cytokine in response to exposure to a vertebrate HMGB B box or biologically active fragment thereof in the absence of a test compound would be expected to be useful for this invention. It is envisioned that the cell that is selected would be important in the etiology ofthe condition to be treated with the inhibitory compound that is being tested. For many conditions, it is expected that the preferred cell is a human macrophage.
  • any method for determining whether the compound inhibits the release ofthe proinflammatory cytokine from the cell would be useful for these embodiments. It is envisioned that the preferred methods are the direct measurement ofthe proinflammatory cytokine, for example, with any of a number of commercially available ELISA assays. However, in some embodiments, the measurement ofthe inflammatory effect of released cytokines may be preferable, particularly when there are several proinflammatory cytokines produced by the test cell. As previously discussed, for many important disorders, the predominant proinflammatory cytokines are TNF, IL-l ⁇ , IL-l ⁇ , MIF or IL-6; particularly TNF.
  • the present invention also features a method of determining whether a compound increases an inflammatory response and/or inflammation.
  • the method comprises combining the compound (test compound) with (a) a cell that releases a proinflammatory cytokine when exposed to a vertebrate HMGB A box or a biologically active fragment thereof, and (b) the HMGB A box or biologically active fragment, and then determining whether the compound increases the release ofthe proinflammatory cytoldne from the cell, as compared to a suitable control.
  • a compound that increases the release ofthe proinflammatory cytokine in this assay is a compound that can be used to increase an inflammatory response and/or inflammation.
  • the HMGB A box or HMGB A box biologically active fragment can be endogenous to the cell or can be introduced into the cell using standard recombinant molecular biology techniques.
  • any cell in which release of a proinflammatory cytokine is normally inhibited in response to exposure to a vertebrate HMGB A box or a biologically active fragment thereof in the absence of any test compound would be expected to be useful for this invention. It is envisioned that the cell that is selected would be important in the etiology ofthe condition to be treated with the inhibitory compound that is being tested. For many conditions, it is expected that the preferred cell is a human macrophage.
  • any method for determining whether the compound increases the release of the proinflammatory cytokine from the cell would be useful for these embodiments. It is envisioned that the preferred methods are the direct measurement ofthe proinflammatory cytokine, for example, with any of a number of commercially available ELISA assays. However, in some embodiments, the measurement ofthe inflammatory effect of released cytokines may be preferable, particularly when there are several proinflammatory cytokines produced by the test cell. As previously discussed, for many important disorders, the predominant proinflammatory cytokines are TNF, IL-l ⁇ , IL-l ⁇ , MIF or IL-6; particularly TNF.
  • HMGBl and Production of HMGBl Mutants The following methods were used to prepare clones and mutants of human HMGBl .
  • Recombinant full length human HMGBl (651 base pairs; GenBank Accession Number U51677) was cloned by PCR amplification from a human brain Quick-Clone cDNA preparation (Clontech, Palo Alto, CA) using the following primers; forward primer: 5' GATGGGCAAAGGAGATCCTAAG 3' (SEQ ID NO:6) and reverse primer: 5* GCGGCCGCTTATTCATCATCATCATCTTC 3' (SEQ ID NO:7).
  • Human HMGBl mutants were cloned and purified as follows.
  • HMGB 1 human Brain Quick-Clone cDNA preparation (Clontech, Palo Alto, CA).
  • the primers used were (forward and reverse, respectively):
  • Carboxy terminus mutant (557 bp): 5' GATGGGCAAAGGAGATCCTAAG 3' (SEQ ID NO:8) and 5' GCGGCCGC TCACTTGCTTTTTTCAGCCTTGAC 3' (SEQ ID NO:9).
  • Amino terminus+B box mutant (486 bp): 5' GAGCATAAGAAGAAGCACCCA 3' (SEQ ID NO: 10) and 5* GCGGCCGC TCACTTGCTTTTTTCAGCCTTGAC 3' (SEQ ID NO: 11).
  • B box mutant (233 bp): 5' AAGTTCAAGGATCCCAATGCAAAG 3' (SEQ ID NO: 12) and 5' GCGGCCGCTCAATATGCAGCTATATCCTTTTC 3' (SEQ ID NO:13).
  • Amino terminus+A box mutant (261 bp): 5' GATGGGCAAAGGAGATCCTAAG 3' (SEQ ID NO: 14) and 5' TCACTTTTTTGTCTCCCCTTTGGG 3' (SEQ ID NO: 15).
  • PCR products were subcloned into pCRII-TOPO vector EcoRI sites using the TA cloning method per manufacturer's instruction (Invitrogen, Carlsbad, CA). After amplification, the PCR product was digested with EcoRI and subcloned into an expression vector with a GST tag pGEX (Pharmacia); correct orientation and positive clones were confirmed by DNA sequencing on both strands. The recombinant plasmids were transformed into protease deficient E.
  • HMGB mutants generated as described above have the following amino acid sequences:
  • a Box mutant MGKGDPKKPTGKMSSYAFFNQTCREEHKKK HPDASV ⁇ FSEFSKKCSERWKTMSAKEKGKFEDMAKADKARYEREMKTYIPP KGET (SEQ ID NO: 21), wherein the A box consists ofthe sequence PTGKMSSYAFFNQTCREEHKKKHPDASNNFSEFSKKCSERWKTMSAKEKGK FEDMAKADKARYEREMKTYIPPKGET (SEQ ID NO:22)
  • a polypeptide generated from a GST vector lacking HMGBl protein was included as a control (containing a GST tag only).
  • DNase I Life Technologies
  • carboxy terminus and B box mutants carboxy terminus and B box mutants
  • benzonase nuclease Novagen, Madison, WI
  • the protein eluates were passed over a polymyxin B column (Pierce, Rockford, IL) to remove any contaminating LPS, and dialyzed extensively against phosphate buffered saline to remove excess reduced glutathione. The preparations were then lyophilized and redissolved in sterile water before use. LPS levels were less than 60 pg/ ⁇ g protein for all ofthe mutants and 300 pg/ ⁇ g for wild type HMG-1, as measured by Limulus amebocyte lysate assay (Bio Whittaker Inc., Walkersville, MD). The integrity of protein was verified by SDS-PAGE. Recombinant rat HMGBl (Wang et al., Science 285: 248-251, 1999) was used in some experiments since it does not have degraded fragments as observed in purified human HMGBl .
  • Murine macrophage-like RAW 264.7 cells (American Type Culture Collection, Rockville, MD) were cultured in RPMI 1640 medium (Lifetime).
  • TNF release was measured by a standard murine fibroblast L929 (ATCC, American Type Culture Collection, Rockville, MD) cytotoxicity bioassay (Bianchi et al., Journal of Experimental Medicine 183:927-936, 1996) with the minimum detectable concentration of 30 pg/ml.
  • Recombinant mouse TNF was obtained from R&D system Inc., (Minneapolis, MN).
  • Murine fibroblast L929 cells were cultured in DMEM (Life Technologies, Grand Island, NY) supplemented with 10% fetal bovine serum (Gemini, Catabasas, CA), penicillin (50 units/ml) and streptomycin (50 ⁇ g/ml) (Life Technologies) in a humidified incubator with 5% CO 2 .
  • HMGBl B box Polyclonal antibodies against HMGBl B box were raised in rabbits (Cocalico Biologicals, Inc., Reamstown, PA) and assayed for titer by immunoblotting. IgG was purified from anti-HMGB 1 antiserum using Protein A agarose according to manufacturer's instructions (Pierce, Rockford, IL). Anti-HMGB 1 B box antibodies were affinity purified using cyanogen bromide activated Sepharose beads (Cocalico Biological, Inc.). Non-immune rabbit IgG was purchased from Sigma (St. Louis, MO). Antibodies detected full length HMGBl and B box in immunoassay, but did not cross react with TNF, IL-1 and IL-6.
  • HMGBl labeling of HMGBl with Na-' 25 I and cell surface binding
  • Purified HMGBl protein (10 ⁇ g) was radiolabeled with 0.2 mCi of carrier- free 125 I (NEN Life Science Products Inc., Boston, MA) using Iodo-beads (Pierce, Rockford, IL) according to the manufacturer's instructions.
  • 125 I-HMGB1 protein was separated from un-reacted 125 I by gel chromatography columns (P6 Micro Bio-Spin Chromatography Columns, Bio-Rad Laboratories, Hercules, CA) previously equilibrated with 300 mM sodium chloride, 17.5 mM sodium citrate, pH 7.0, and 0.1%) bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • TNF knock out mice were obtained from Amgen (Thousand Oaks, CA) and were on a B6xl29 background. Age-matched wild-type B6xl29 mice were used as a control for the studies. Mice were bred in-house at the University of Florida specific pathogen-free transgenic mouse facility (Gainesville, FL) and were used at 6-8 weeks of age.
  • mice Male 6-8 week old Balb/c and C3H/HeJ mice were purchased from Harlen Sprague-Dawley (Indianapolis, IN) and were allowed to acclimate for 7 days before use in experiments. All animals were housed in the North Shore University Hospital Animal Facility under standard temperature, and a light and dark cycle.
  • Cecal ligation and puncture was performed as described previously (Fink and Heard, J. Surg. Res. 49:186-196, 1990; Wichmann et al, Crit. Care Med. 26:2078-2086, 1998; and Remick et al, Shock 4:89-95, 1995). Briefly, Balb/c mice were anesthetized with 75 mg/kg ketamine (Fort Dodge, Fort Dodge, Iowa) and 20 mg/kg of xylazine (Bohringer Ingelheim, St. Joseph, MO) intramuscularly. A midline incision was performed, and the cecum was isolated. A 6-0 prolene suture ligature was placed at a level 5.0 mm from the cecal tip away from the ileocecal valve.
  • the ligated cecal stump was then punctured once with a 22-gauge needle, without direct extrusion of stool.
  • the cecum was then placed back into its normal intra-abdominal position.
  • the abdomen was then closed with a running suture of 6-0 prolene in two layers, peritoneum and fascia separately to prevent leakage of fluid.
  • All animals were resuscitated with a normal saline solution administered sub- cutaneously at 20 ml/kg of body weight.
  • Each mouse received a subcutaneous injection of imipenem (0.5 mg/mouse) (Primaxin, Merck & Co., Inc., West Point, PA) 30 minutes after the surgery. Animals were then allowed to recuperate. Mortality was recorded for up to 1 week after the procedure; survivors were followed for 2 weeks to ensure no late mortalities had occurred.
  • mice were injected intraperitoneally with 20 mg D-galactosamine-HCL (Sigma)/mouse (in 200. ⁇ l PBS) and 0.1 or 1 mg of either HMBG1 B box or vector protein (in 200 ⁇ l PBS). Mortality was recorded daily for up to 72 hours after injection; survivors were followed for 2 weeks, and no later deaths from B box toxicity were observed.
  • Spleen bacteria were recovered as described previously (Villa et al., J. Endotoxin Res. 4:197-204, 1997). Spleens were removed using sterile technique and homogenized in 2 ml of PBS. After serial dilutions with PBS, the homogenate was plated as 0.15 ml aliquots on tryptic soy agar plates (Difco, Detroit, MI) and CFU were counted after overnight incubation at 37°C.
  • HMGBl has 2 folded DNA binding domains (A and B boxes) and a negatively-charged acidic carboxyl tail.
  • a and B boxes DNA binding domains
  • carboxyl tail a negatively-charged acidic carboxyl tail.
  • mutants of human HMGBl were made by polymerase chain reaction (PCR) using specific primers as described herein, and the mutant proteins were expressed using a glutathione S-transferase (GST) gene fusion system (Pharmacia Biotech, Piscataway, NJ) in accordance with the manufacturer's instructions. Briefly, DNA fragments, made by PCR methods, were fused to GST fusion vectors and amplified in E. coli. The expressed HMGBl protein and HMGBl mutants were then isolated using a GST affinity column.
  • GST glutathione S-transferase
  • RAW 264.7 cells were cultured in RPMI 1640 medium (Life Technologies, Grand Island NY) supplemented with 10% fetal bovine serum (Gemini, Catabasas, CA), penicillin and streptomycin (Life Technologies). Polymyxin (Sigma, St. Louis, MO) was added at 100 units/ml to suppress the activity of any contaminating LPS. Cells were incubated with 1 ⁇ g/ml of full length (wild-type) HMGBl and each HMGBl mutant protein in Opti- MEM I medium for 8 hours.
  • TNF released from the cells was measured by a standard murine fibroblast L929 (ATCC) cytotoxicity bioassay (Bianchi et al., supra) with the minimum detectable concentration of 30 pg/ml.
  • HMGBl and carboxy 1-truncated HMGBl significantly stimulated TNF release by monocyte cultures (murine macrophage-like RAW 264.7 cells).
  • the B box was a potent activator of monocyte TNF release. This stimulating effect ofthe B box was specific, because A box only weakly activated TNF release.
  • HMGBl B box varying amounts of HMGBl B box were evaluated for the effects on TNF, IL-1B, and IL-6 production in murine macrophage-like RAW 264.7 cells.
  • RAW 264.7 cells were stimulated with B box protein at 0-10 ⁇ g/ml, as indicated in FIGS. 2A-2C for 8 hours.
  • Conditioned media were harvested and measured for TNF, IL-l ⁇ and IL-6 levels. TNF levels were measured as described herein, and IL-l ⁇ and IL-6 levels were measured using the mouse IL-l ⁇ and IL-6 enzyme-linked immunosorbent assay (ELISA) kits (R&D System Inc., Minneapolis, MN) and N>5 for all experiments.
  • ELISA enzyme-linked immunosorbent assay
  • FIGS. 2A-2C The results ofthe studies are shown in FIGS. 2A-2C.
  • TNF release from RAW 264.7 cells increased with increased amounts of B box administered to the cells.
  • FIG. 2B addition of 1 ⁇ g/ml or 10 ⁇ g/ml of B box resulted in increased release of IL-l ⁇ from RAW 264.7 cells.
  • FIG. 2C IL-6 release from RAW 264.7 cells increased with increased amounts of B box administered to the cells. The kinetics of B box-induced TNF release were also examined.
  • TNF release and TNF mRNA expression were measured in RAW 264.7 cells induced by B box polypeptide or GST tag polypeptide only used as a control (vector) (10 ⁇ g/ml) for 0 to 48 hours.
  • RNAzol B RNAzol B method in accordance with the manufacturer's instructions (Tel-Test "B", Inc., Friendswood, TX). TNF (287 bp) was measured by RNase protection assay (Ambion, Austin, TX). Equal loading and the integrity of RNA was verified by ethidium bromide staining ofthe RNA sample on an agarose-formaldehyde gel. The results ofthe RNase protection assay are shown in FIG. 2D. As shown in FIG. 2D, B box activation of monocytes occurred at the level of gene transcription, because TNF mRNA was increased significantly in monocytes exposed to B box protein (FIG. 2B).
  • TNF mRNA expression was maximal at 4 hours and decreased at 8 and 24 hours.
  • the vector only control (GST tag) showed no effect on TNF mRNA expression.
  • a similar study was carried out measuring TNF protein released from RAW 264.7 cells 0, 4, 8, 24, 32 or 48 hours after administration of B box or vector only (GST tag), using the L929 cytotoxicity assay described herein. Compared to the control (medium only), B box treatment stimulated TNF protein expression (FIG. 2E) and vector alone (FIG. 2F) did not. Data are representative of three separate experiments. Together these data indicate that the HMGB 1 B box domain has cytokine activity and is responsible for the cytokine stimulating activity of full length HMGB 1.
  • HMGBl B box dose-dependently stimulated release of TNF, IL-l ⁇ and IL-6 from monocyte cultures (FIGS. 2A-2C), in agreement with the inflammatory activity of full length HMGBl (Andersson et al., J. Exp. Med. 192: 565-570, 2000).
  • these studies indicate that maximum TNF protein release occurred within 8 hours (FIG. 2E).
  • This delayed pattern of TNF release is similar to TNF release induced by HMGBl itself, and is significantly later than the kinetics of TNF induced by LPS (Andersson et al, supra).
  • Example 4 The First 20 Amino Acids ofthe HMGBl B Box Stimulate TNF Activity
  • the TNF-stimulating activity ofthe HMGBl B box was further mapped. This study was carried out as follows. Fragments ofthe B box were generated using synthetic peptide protection techniques, as described herein. Five HMGBl B box fragments (from SEQ ID NO:20), containing amino acids 1-20, 16-25, 30-49, 45-64, or 60-74 ofthe HMGBl B box were generated, as indicated in FIG. 3. RAW 264.7 cells were treated with B box (1 ⁇ g/ml) or a synthetic peptide fragment ofthe B box (10 ⁇ g/ml), as indicated in FIG.
  • TNF-stimulating activity was retained by a synthetic peptide corresponding to amino acids 1-20 ofthe HMGBl B box of SEQ ID NO:20 (flcdpnaplcrlpsafflfcse; SEQ ID NO :23).
  • the TNF stimulating activity of the 1 -20- mer was less potent than either the full length synthetic B box (1-74-mer), or full length HMGBl, but the stimulatory effects were specific because the synthetic 20- mers for amino acid fragments containing 16-25, 30-49, 45-64, or 60-74 ofthe HMGBl B box did not induce TNF release.
  • This B box fragment can be used in the same manner as a polypeptide encoding a full length B box polypeptide, for example, to stimulate release of a proinflammatory cytokine, or to treat a condition in a patient characterized by activation of an inflammatory cytokine cascade.
  • HMGBl A box only weakly induced TNF production, as shown in FIG. 1, the ability of HMGBl A box to act as an antagonist of HMGBl activity was evaluated. This study was carried out as follows. Sub-confluent RAW 264.7 cells in 24-well dishes were treated with HMGBl (1 ⁇ g/ml) and 0, 5, 10, or 25 ⁇ g/ml of A box for 16 hours in Opti-MEM I medium in the presence of polymyxin B (100 units/ml).
  • the TNF-stimulating activity (assayed using the L929 cytotoxicity assay described herein) in the sample receiving no A box was expressed as 100%, and the inhibition by A box was expressed as percent of HMGBl alone.
  • the results ofthe effect of A box on TNF release from RAW 264.7 cells is shown in FIG. 4A.
  • Antagonism of full length HMGBl activity by HMGBl was also determined by measuring TNF release from RAW 264.7 macrophage cultures stimulated by co-addition of A box with full length HMGBl.
  • RAW 264.7 macrophage cells ATCC were seeded into 24-well tissue culture plates and used at 90% confluence. The cells were treated with HMGBl, and/or A boxes as indicated for 16 hours in Optimum I medium (Life Technologies, Grand Island, NY) in the presence of polymyxin B (100 units/ml, Sigma, St. Louis, MO) and supematants were collected for TNF measurement (mouse ELISA kit from R&D System Inc, Minneapolis, MN).
  • FIG. 4B is a histogram ofthe effect of HMGBl (HMG-1), alone, A box alone, Vector (control) alone, HMGBl in combination with A box, and HMGBl in combination with vector. As shown in FIG. 4B, HMGBl A box significantly attenuated the TNF stimulating activity of full length HMGBl.
  • Example 7 HMGBl A Box Protein Inhibits HMGBl Cytokine Activity by Binding to It
  • HMGBl A box acts as an antagonist by displacing HMGBl binding
  • 125 I-labeled-HMGBl was added to macrophage cultures and binding was measured at 4°C after 2 hours. Binding assays in RAW 264.7 cells were performed as described herein. 125 I-HMGB1 binding was measured in RAW 264.7 cells plated in 24-well dishes for the times indicated in FIG. 5A. Specific binding shown equals total cell-associated 125 I-HMGB1 (CPM/well) minus cell associated CPM/well in the presence of 5,000 fold molar excess of unlabeled HMGBl.
  • FIG. 5 A is a graph ofthe binding of 125 I-HMGB1 over time. As shown in FIG. 5 A,
  • HMGBl exhibited saturable first order binding kinetics. The specificity of binding was assessed as described in Example 1.
  • FIG. 5B is a histogram ofthe cell surface binding of 125 I-HMGB1 in the absence of unlabeled HMGBl or HMGBl A box, or in the presence of 5,000 molar excess of unlabeled HMGBl or HMGBl A box, measured as a percent ofthe total CPM/well.
  • Total equals counts per minutes (CPMVwell of cell associated 125 I-HMGB1 in the absence of unlabeled HMGBl or A box for 2 hours at 4°C.
  • HMGBl or A box equals CPM/well of cell-associated 125 I-HMGB1 in the presence of 5,000 molar excess of unlabeled HMGBl or unlabeled A box. The data are expressed as the percent of total counts obtained in the absence of unlabeled HMGBl proteins (2,382,179 CPM/well).
  • Example 8 Inhibition of Full Length HMGBl and HMGBl B Box Cytokine Activity by Anti-B Box Polyclonal Antibodies.
  • HMGBl B box antibodies The ability of antibodies directed against the HMGBl B box to modulated the effect of full length or HMGB 1 B box was also assessed.
  • Affinity purified antibodies directed against the HMGBl B box (B box antibodies) were generated as described herein and using standard techniques.
  • To assay the effect ofthe antibodies on HMGBl -induced or HMGBl B box-induced TNF release from RAW 264.7 cells sub-confluent RAW 264.7 cells in 24-well dishes were treated with HMG-1 (1 ⁇ g/ml) or HMGB 1 B box (10 ⁇ g/ml) for 10 hours with or without anti-B box antibody (25 ⁇ g/ml or 100 ⁇ g/ml antigen affinity purified, Cocalico Biologicals, Inc., Reamstown, PA) or non-immune IgG (25 ⁇ g/ml or 100 ⁇ g/ml; Sigma) added.
  • FIG. 6 is a histogram of TNF released by RAW 264.7 cells administered nothing, 1 ⁇ g/ml of HMGB 1 , 1 ⁇ g/ml of HMGBl plus 25 ⁇ g/ml of anti-B box antibody, 1 ⁇ g/ml of HMGBl plus 25 ⁇ g/ml of IgG (control), 10 ⁇ g/ml of B-box, 10 ⁇ g/ml of B-box plus 100 ⁇ g/ml of anti-B box antibody or 10 ⁇ g/ml of B-box plus 100 ⁇ g/ml of IgG (control).
  • the amount of TNF released from the cells induced by HMGBl alone (without addition of B box antibodies) was set as 100%, and the data shown in FIG. 6 are the results of 3 independent experiments. As shown in FIG. 6, affinity purified antibodies directed against the HMGBl B box significantly inhibited TNF release induced by either full length HMGBl or the HMGBl B box. These results indicate that such an antibody can be used to modulate HMGBl function.
  • mice (20-25 grams, male, Harlan Sprague-Dawley, Indianapolis, IN) were intraperitoneally injected with D-gal (20 mg) (Sigma, St. Louis, Missouri) and B box (0.1 mg/ml/mouse or 1 mg/ml/mouse) or GST tag (vector; 0.1 mg/ml/mouse or 1 mg/ml/mouse), as indicated in Table 1. Survival of the mice was monitored up to 7 days to ensure no late death occurred. The results of this study are shown in Table 1. Table 1: Toxicity of HMGBl B box on D-galactosamine-sensitized Balb/c Mice
  • mice treated with comparable preparations ofthe purified GST vector protein devoid of B box were lethal to D- galactosamine-sensitized mice in a dose-dependent mamier. In all instances in which death occurred, it occurred within 12 hours. Lethality was not observed in mice treated with comparable preparations ofthe purified GST vector protein devoid of B box.
  • Example 10 Histology of D-galactosamine-sensitized Balb/c Mice or C3H/HeJ Mice Administered HMGBl B Box Protein
  • mice D-galactosamine-sensitized Balb/c mice.
  • Mice (3 per group) received D-gal (20 mg/mouse) plus B box or vector (1 mg/mouse) intraperitoneally for 7 hours and were then sacrificed by decapitation. Blood was collected, and organs (liver, heart, kidney and lung) were harvested and fixed in 10% formaldehyde. Tissue sections were prepared with hematoxylin and eosin staining for histological evaluation (Criterion Inc., Vancouver, Canada). The results of these studies are shown in FIGS.
  • FIG. 7A-7J which are scanned images of hematoxylin and eosin stained kidney sections (FIG. 7A), myocardium sections (FIG. 7C), lung sections (FIG. 7E), and liver sections (FIGS. 7G and 71) obtained from an untreated mouse and kidney sections (FIG. 7B), myocardium sections (FIG. 7D), lung sections (FIG. 7F), and liver sections (FIGS. 7H and 7J) obtained from mice treated with the HMGB 1 B box.
  • B box treatment caused no abnormality in kidneys (FIGS. 7A and 7B) and lungs (FIGS. 7E and 7F).
  • mice had some ischemic changes and loss of cross striation in myocardial fibers in the heart (FIGS. 7C and 7D as indicated by the arrow in FIG. 7D).
  • Liver showed most ofthe damage by the B box as illustrated by active hepatitis (FIGS. 7G-7J).
  • FIG. 7J hepatocyte dropouts are seen surrounded by accumulated polymorphonuclear leukocytes.
  • the arrows in FIG. 7J point to the sites of polymorphonuclear accumulation (dotted) or apoptotic hepatocytes (solid).
  • Administration of HMGBl B box in vivo also stimulated significantly increased serum levels of IL-6 (315+93 vs.20+7 pg/ml, B box vs. control, p ⁇ 0.05) and IL-l ⁇ (15+3 vs. 4+1 pg/ml, B box vs. control, p ⁇ 0.05).
  • TNF-KO TNF knock-out mice
  • wild-type controls B6xl29 strain sensitized with D- galactosamine (20 mg/mouse) and exposed to B box (1 mg/mouse, injected intraperitoneally).
  • the B box was highly lethal to the wild-type mice (6 dead out of nine exposed) but lethality was not observed in the TNF-KO mice treated with B box (0 dead out of 9 exposed, p ⁇ 0.05 v. wild type).
  • FIG. 8 shows the results of this study in a graph that illustrates the levels of HMGBl in mice 0 hours, 8 hours, 18 hours, 24 hours, 48 hours, and 72 hours after subjection to CLP.
  • serum HMGBl levels were not significantly increased for the first eight hours after cecal perforation, then increased significantly after 18 hours (FIG. 8).
  • Increased serum HMGBl remained at elevated plateau levels for at least 72 hours after CLP, a kinetic profile that is quite similar to the previously-described, delayed HMGB 1 kinetics in endotoxemia (Wang et al., supra). This temporal pattern of HMGBl release corresponded closely to the development of signs of sepsis in the mice.
  • mice were subjected to cecal perforation and treated by administration of A box beginning 24 hours after the onset of sepsis.
  • CLP was performed on male Balb/c mice as described herein. Animals were randomly grouped, with 15-25 mice per group.
  • the HMGBl A box (60 or 600 ⁇ g/mouse each time) or vector (GST tag, 600 ⁇ g/mouse) alone was administered intraperitoneally twice daily for 3 days beginning 24 hours after CLP. Survival was monitored twice daily for up to 2 weeks to ensure no late death occurred. The results of this study are illustrated in FIG.
  • the rescuing effects ofthe HMGBl A box in this sepsis model were A box dose-dependent; animals treated with 600 ⁇ g/mouse of A box were observed to be significantly more alert, active, and to resume feeding behavior as compared to either control animals treated with vector-derived preparations, or to animals treated with only 60 ⁇ g A box. The latter animals remained gravely ill, with depressed activity and feeding for several days, and most died.
  • FIG. 10 A is a graph ofthe survival of septic mice treated with either a control antibody or an anti- HMGB 1 antibody.
  • CFU the aerobic colony forming units
  • anti-HMGB 1 B box antibodies were evaluated for their ability to rescue LPS-induced septic mice.
  • Male Balb/c mice (20-25 gm, 26 per group) were treated with an LD75 dose of LPS (15 mg/kg) injected intraperitoneally (IP).
  • Anti-HMGB 1 B box or non-immune rabbit serum (0.3 ml per mouse each time, IP) was given at time 0, +12 hours and +24 hours after LPS administration. Survival of mice was evaluated over time. The results of this study are shown in FIG. 10B, which is a graph ofthe survival of septic mice administered anti-HMGB 1 B box antibodies or non-immune serum. As shown in FIG. 10B, anti-HMGB 1 B box antibodies improved survival ofthe septic mice.
  • Example 14 Inhibition of HMGBl Signaling Pathway Using an Anti-RAGE Antibody
  • HMGBl HMG-1; 1 ⁇ g/ml
  • LPS 0.1 ⁇ g/ml
  • HMGBl B box B Box; 1 ⁇ g/ml
  • anti-RAG ⁇ antibody 25 ⁇ g/ml
  • non-immune IgG 25 ⁇ g/ml
  • supematants were collected for TNF measurement using the L929 cytotoxicity assay described herein.
  • IgG purified polyclonal anti- RAG ⁇ antibody Catalog No.
  • FIG. 11 A is a histogram ofthe effects of HMGB 1, LPS, or HMGB 1 B box in the presence of anti-RAG ⁇ antibodies or non-immune IgG (control) on TNF release from RAW 264.7 cells.
  • anti-RAG ⁇ antibody significantly inhibited HMGBl B box-induced TNF release. This suppression was specific, because anti-RAG ⁇ did not significantly inhibit LPS-stimulated TNF release.
  • the maximum inhibitory effect of anti- RAGE decreased HMG-1 signaling by only 40%, suggesting that other signal transduction pathways may participate in HMGBl signaling.
  • HMGB 1 or HMGB 1 B box were transiently co-transfected with an expression plasmid encoding a murine MyD 88-dominant-negative (DN) mutant (corresponding to amino acids 146-296), or empty vector, plus a luciferase reporter plasmid under the control ofthe NF- ⁇ B -dependent ELAM promoter, as described by Means et al. (J. Immunol. 166:4074-4082, 2001).
  • DN MyD 88-dominant-negative
  • HMGBl full-length HMGBl (100 ng/ml), or purified HMGBl B box (10 ⁇ g/ml), for 5 hours.
  • Cells were then harvested and luciferase activity was measured, using standard methods. All transfections were performed in triplicate, repeated at least three times, and a single representative experiment is shown in FIG. 1 IB.
  • HMGBl stimulated luciferase activity in samples that were not co-transfected with the MyD 88 dominant negative, and the level of stimulation was decreased in samples that were co-transfected with the MyD 88 dominant negative. This effect was also observed in samples administered HMGB B box.

Abstract

Compositions and methods are disclosed for inhibiting the release of a proinflammatory cytokine from a cell, and for inhibiting an inflammatory cytokine cascade in a patient. The compositions comprise an HMGB A box, and/or an antibody preparation that specifically binds to an HMGB B box, and/or an inhibitor of TNF biological activity. The methods comprise treating a cell or a patient with sufficient amounts of the composition to inhibit the release of the proinflammatory cytokine, or to inhibit the inflammatory cytokine cascade.

Description

USE OF HMGB FRAGMENTS AS ANTI-INFLAMMATORY AGENTS
RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Nos. 60/427,841 and 60/427,846, both of which were filed on November 20, 2002. The entire teachings of both applications are incorporated herein by reference.
BACKGROUND OF THE INVENTION
Inflammation is often induced by proinflammatory cytokines, such as tumor necrosis factor (TNF), inter leukin (IL)-l , IL-lβ, IL-6, platelet-activating factor (PAF), macrophage migration inhibitory factor (MIF), and other compounds. These proinflammatory cytokines are produced by several different cell types, most importantly immune cells (for example, monocytes, macrophages and neutrophils), but also non-immune cells such as fϊbroblasts, osteoblasts, smooth muscle cells, epithelial cells, and neurons. These proinflammatory cytokines contribute to various disorders during the early stages of an inflammatory cytokine cascade. Inflammatory cytokine cascades contribute to deleterious characteristics, including inflammation and apoptosis, of numerous disorders. Included are disorders characterized by both localized and systemic reactions, including, without limitation, diseases involving the gastrointestinal tract and associated tissues (such as appendicitis, peptic, gastric and duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute and ischemic colitis, diverticulitis, epiglottitis, achalasia, cholangitis, cholecystitis, coeliac disease, hepatitis, Crohn's disease, enteritis, and Whipple's disease); systemic or local inflammatory diseases and conditions (such as asthma, allergy, anaphylactic shock, immune complex disease, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, hyperpyrexia, eosinophilic granuloma, granulomatosis, and sarcoidosis); diseases involving the urogenital system and associated tissues (such as septic abortion, epididymitis, vaginitis, prostatitis, and urethritis); diseases involving the respiratory system and associated tissues (such as bronchitis, emphysema, rhinitis, cystic fibrosis, pneumonitis, adult respiratory distress syndrome, pneumoultramicroscopicsilicovolcanoconiosis, alvealitis, bronchiolitis, pharyngitis, pleurisy, and sinusitis); diseases arising from infection by various viruses (such as influenza, respiratory syncytial virus, HIN, hepatitis B virus, hepatitis C virus and herpes), bacteria (such as disseminated bacteremia, Dengue fever), fungi (such as candidiasis) and protozoal and multicellular parasites (such as malaria, filariasis, amebiasis, and hydatid cysts); dermatological diseases and conditions ofthe skin (such as burns, dermatitis, dermatomyositis, sunburn, urticaria warts, and wheals); diseases involving the cardiovascular system and associated tissues (such as vasulitis, angiitis, endocarditis, arteritis, atherosclerosis, restenosis, thrombophlebitis, pericarditis, congestive heart failure, myocarditis, myocardial ischemia, periarteritis nodosa, and rheumatic fever); diseases involving the central or peripheral nervous system and associated tissues (such as Alzheimer's disease, meningitis, encephalitis, multiple sclerosis, cerebral infarction, cerebral embolism, Guillame-Barre syndrome, neuritis, neuralgia, spinal cord injury, paralysis, and uveitis); diseases ofthe bones, joints, muscles and connective tissues (such as the various arthritides and arthralgias, osteomyelitis, fasciitis, Paget's disease, gout, periodontal disease, rheumatoid arthritis, and synovitis); other autoimmune and inflammatory disorders (such as myasthenia gravis, thryoiditis, systemic lupus erythematosus, Goodpasture's syndrome, Behcets's syndrome, allograft rejection, graft- versus-host disease, Type I diabetes, ankylosing spondylitis, Berger' s disease, and Retier's syndrome); as well as various cancers, tumors and proliferative disorders (such as Hodgkins disease); and, in any case the inflammatory or immune host response to any primary disease. The early proinflammatory cytokines (e.g., TΝF, IL-1, etc.) mediate inflammation, and induce the late release of high mobility group box 1 (HMGB1) (also known as HMG-1 and HMG1), a protein that accumulates in serum and mediates delayed lethality and further induction of early proinflammatory cytokines.
HMGBl was first identified as the founding member of a family of DNA- binding proteins termed high mobility group box (HMGB) proteins that are critical for DNA structure and stability. It was identified nearly 40 years ago as a ubiquitously expressed nuclear protein that binds double-stranded DNA without sequence specificity.
HMGBl binding bends DNA to promote formation and stability of nucleoprotein complexes that facilitate gene transcription of glucocorticoid receptors and RAG recombinase. The HMGBl molecule has three domains: two DNA binding motifs termed HMGB A and HMGB B boxes, and an acidic carboxyl terminus. The two HMGB boxes are highly conserved 80 amino acid, L-shaped domains. HMGB boxes are also expressed in other transcription factors including the RNA polymerase I transcription factor human upstream-binding factor and lymphoid-specific factor.
Recent evidence has implicated HMGBl as a cytokine mediator of inflammatory conditions. For example, HMGBl has been implicated as a cytokine mediator of delayed lethality in endotoxemia. That work demonstrated that bacterial endotoxin (lipopolysaccharide (LPS)) activates monocytes/macrophages to release HMGBl as a late response to activation, resulting in elevated serum HMGBl levels that are toxic. Antibodies against HMGB 1 prevent lethality of endotoxin even when antibody administration is delayed until after the early cytokine response. Like other proinflammatory cytokines, HMGBl is a potent activator of monocytes. Intratracheal application of HMGBl causes acute lung injury, and anti-HMGBl antibodies protect against endotoxin-induced lung edema. Serum HMGBl levels are elevated in critically ill patients with sepsis or hemorrhagic shock, and levels are significantly higher in non-survivors as compared to survivors.
HMGBl has also been implicated as a ligand for RAGE, a multi-ligand receptor ofthe immunoglobulin superfamily. RAGE is expressed on endothelial cells, smooth muscle cells, monocytes, and nerves, and ligand interaction transduces signals through MAP kinase, P21 ras, andNF-κB. The delayed kinetics of HMGBl appearance during endotoxemia makes it a potentially good therapeutic target, but little is known about the molecular basis of HMGBl signaling and toxicity. Therefore, it would be useful to identify characteristics of HMGBl proinflammatory activity, particularly the active domain(s) responsible for this activity, and any inhibitory effects of other domains.
SUMMARY OF THE INVENTION
The present invention is based on the discoveries that (1) the HMGB A box serves as a competitive inhibitor of HMGB proinflammatory action, (2) the HMGB B box has the predominant proinflammatory activity of HMGB, and (3) combination therapies involving agents that inhibit HMGB biological activity and agents that inhibit TNF biological activity can be used for the treatment of conditions characterized by activation ofthe inflammatory cytokine cascade. Agents that inhibit HMGB biological activity include the HMGB A box, which serves as a competitive inhibitor of HMGB proinflammatory action, and antibodies to HMGB, for example, the HMGB B box.
Accordingly, in one embodiment, the invention is a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof, or an A box biologically active fragment or variant thereof, which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, wherein the HMGB A box is selected from the group consisting of an
HMG1L5 (formerly HMG1L10) A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RP11-395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box. In one embodiment, the polypeptide can be in a pharmaceutically acceptable carrier. In another embodiment, the invention is a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, wherein the antibodies can inhibit release of a proinflammatory cytokine from a cell treated with HMGB, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23. In one embodiment, the antibodies can be in a pharmaceutically acceptable carrier.
In still another embodiment, the invention is a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, or a B box biologically active fragment or variant thereof, but not comprising a full length HMGB, wherein the polypeptide can cause release of a proinflammatory cytokine from a cell, and wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23. In one embodiment, the polypeptide can be in a pharmaceutically acceptable carrier. In other embodiments, the invention comprises vectors encoding the polypeptides described above.
In still another embodiment, the invention is a method of inhibiting release of a proinflammatory cytokine from a mammalian cell, the method comprising treating the cell with an amount of a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (fonnerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23. In another embodiment, the invention is a method of inhibiting release of a proinflammatory cytokine from a mammalian cell, the method comprising treating the cell with a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof, or an A box biologically active fragment or variant thereof, which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in an amount sufficient to inhibit release ofthe proinflammatory cytokine from the cell, wherein the HMGB A box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RP11- 395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box. In one embodiment, the cell can be treated with a vector encoding a polypeptide comprising the A box polypeptide, A box biologically active fragment, or variant thereof.
In another embodiment, the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, in an amount sufficient to inhibit the inflammatory cytokine cascade, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
In another embodiment, the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof, or an A box biologically active fragment or variant thereof, which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, in an amount sufficient to inhibit release ofthe proinflammatory cytokine from the cell, wherein the HMGB A box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl l-395A23, an HMGlL9 A box, an LOC122441 B box, an LOCI 39603 A box, and an HMG1L8 A box.
In still another embodiment, the invention is a method of stimulating the release of a proinflammatory cytokine from a cell comprising treating the cell with a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, or a B box biologically active fragment thereof, but not comprising a full length HMGB, in an amount sufficient to stimulate the release ofthe proinflammatory cytokine from the cell, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1- 395A23. In one embodiment, the cell can be treated with a vector encoding a polypeptide comprising the B box polypeptide, B box biologically active fragment, or variant thereof.
In still another embodiment, the invention is a method for effecting weight loss or treating obesity in a patient, comprising administering to the patient an effective amount of a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, or a B box biologically active fragment or variant thereof, but not comprising a full length HMGB polypeptide, in an amount sufficient to stimulate the release of a proinflammatory cytokine from a cell, wherein the HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
In another embodiment, the invention is a method of determining whether a compound inhibits inflammation, comprising combining the compound with a) a cell that releases a proinflammatory cytokine when exposed to a high mobility group box protein (HMGB) B box or a biologically active fragment thereof; and b) the HMGB B box or biologically active fragment thereof, wherein said HMGB B box is selected from the group consisting of an HMG1L5 (formerly HMG1L10) B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23; then determining whether the compound inhibits the release of the proinflammatory cytokine from the cell.
In yet another embodiment, the invention is a pharmaceutical composition comprising a polypeptide comprising a high mobility group box (HMGB) A box, or a fragment or variant thereof, that can inhibit release of a proinflammatory cytokine from a cell treated with a high mobility group box (HMGB) protein and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide, in a pharmaceutically acceptable carrier. The HMGB A box is preferably a vertebrate HMGB A box, for example, a mammalian HMGB A box, more preferably, a mammalian HMGBl A box, for example, a human HMGBl A box, and most preferably, the HMGBl A box comprising or consisting ofthe sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57. In another embodiment, the invention is a pharmaceutical composition comprising an antibody that binds an HMGB polypeptide or a biologically active fragment thereof, for example, an HMGB B box polypeptide or biologically active fragment thereof, and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab,
CDP870, CDP571, Lenercept, and Thalidomide, in a pharmaceutically acceptable carrier.
In still another embodiment, the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade comprising administering to the patient a composition comprising a polypeptide comprising a high mobility group box (HMGB) A box or a fragment or variant thereof that can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide.
In still another embodiment, the invention is a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade comprising administering to the patient a composition comprising an antibody that binds an HMGB polypeptide or a biologically active fragment thereof, for example, an HMGB B box polypeptide or a biologically active fragment thereof, and an agent that inhibits TNF biological activity, where the agent is selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is a schematic representation of HMGBl mutants and their activity in TNF release (pg/ml).
FIG. 2A is a histogram showing the effect of 0 μg/ml, 0.01 μg/ml, 0.1 μg/ml, 1 μg/ml or 10 μg/ml of B box on TNF release (pg/ml) in RAW 264.7 cells. FIG. 2B is a histogram showing the effect of 0 μg/ml, 0.01 μg/ml, 0.1 μg/ml, 1 μg/ml or 10 μg/ml of B box on IL-lβ release (pg/ml) in RAW 264.7 cells.
FIG. 2C is a histogram showing the effect of 0 μg/ml, 0.01 μg/ml, 0.1 μg/ml, 1 μg/ml or 10 μg/ml of B box on IL-6 release (pg/ml) in RAW 264.7 cells. FIG. 2D a scanned image of a blot of an RNAse protection assay, showing the effect of B box (at 0 hours, 4 hours, 8 hours, or 24 hours after administration) or vector alone (at 4 hours after administration) on TNF mRNA expression in RAW 264.7 cells.
FIG. 2E is a histogram ofthe effect of HMGBl B box on TNF protein release (pg/ml) from RAW 264.7 cells at 0 hours, 4 hours, 8 hours, 24 hours, 32 hours or 48 hours after administration.
FIG. 2F is a histogram ofthe effect of vector on TNF protein release (pg/ml) from RAW 264.7 cells at 0 hours, 4 hours, 8 hours, 24 hours, 32 hours or 48 hours after administration. FIG. 3 is a schematic representation of HMGB 1 B box mutants and their activity in TNF release (pg/ml).
FIG. 4A is a graph ofthe effect of 0 μg/ml, 5 μg/ml, 10 μg/ml, or 25 μg/ml of HMG1 A box protein on the release of TNF (as a percent of HMGBl mediated TNF release alone) from RAW 264.7 cells. FIG. 4B is a histogram of the effect of HMGB 1 (0 or 1.5 μg/ml), HMGB 1 A box (0 or 10 μg/ml), or vector (0 or 10 μg/ml), alone, or in combination, on the release of TNF (as a percent of HMGBl mediated TNF release alone) from RAW 264.7 cells.
FIG. 5A is a graph of binding of 125I-HMGB1 binding to RAW 264.7 cells (CPM/well) over time (minutes).
FIG. 5B is a histogram ofthe binding of 125I-HMGB1 in the absence of unlabeled HMGBl or HMGBl A box for 2 hours at 4°C (Total), or in the presence of 5,000 molar excess of unlabeled HMGBl (HMGBl) or A box (A box), measured as a percent ofthe total CPM/well. FIG. 6 is a histogram ofthe effects of HMGB 1 (HMG-1 ; 0 μg/ml or 1 μg/ml) or HMGBl B box (B Box; 0 μg/ml or 10 μg/ml), alone or in combination with anti-B box antibody (25 μg/ml or 100 μg/ml) or IgG (25 μg/ml or 100 μg/ml) on TNF release from RAW 264.7 cells (expressed as a percent of HMGBl mediated TNF release alone).
FIG. 7A is a scanned image of a hematoxylin and eosin stained kidney section obtained from an untreated mouse.
FIG. 7B is a scanned image of a hematoxylin and eosin stained kidney section obtained from a mouse administered HMGBl B box.
FIG. 7C is a scanned image of a hematoxylin and eosin stained myocardium section obtained from an untreated mouse. FIG. 7D is a scanned image of a hematoxylin and eosin stained myocardium section obtained from a mouse administered HMGBl B box.
FIG. 7E is a scanned image of a hematoxylin and eosin stained lung section obtained from an untreated mouse.
FIG. 7F is a scanned image of a hematoxylin and eosin stained lung section obtained from a mouse administered HMGB 1 B box.
FIG. 7G is a scanned image of a hematoxylin and eosin stained liver section obtained from an untreated mouse.
FIG. 7H is a scanned image of a hematoxylin and eosin stained liver section obtained from a mouse administered HMGBl B box. FIG. 71 is a scanned image of a hematoxylin and eosin stained liver section
(high magnification) obtained from an untreated mouse.
FIG. 7J is a scanned image of a hematoxylin and eosin stained liver section (high magnification) obtained from a mouse administered HMGBl B box.
FIG. 8 is a graph ofthe level of HMGBl (ng/ml) in mice subjected to cecal ligation and puncture (CLP) over time (hours).
FIG. 9 is a graph ofthe effect of HMGB A Box (60 μg/mouse or 600 μg/mouse) or no treatment on survival of mice over time (days) after cecal ligation and puncture (CLP).
FIG. 10A is a graph ofthe effect of anti-HMGBl antibody (dark circles) or no treatment (open circles) on survival of mice over time (days) after cecal ligation and puncture (CLP). FIG. 1 OB is a graph ofthe effect of anti-HMGBl B box antiserum (■) or no treatment (*) on the survival (days) of mice administered lipopolysaccharide (LPS).
FIG. 11A is a histogram ofthe effect of anti-RAGE antibody or non-immune IgG on TNF release from RAW 264.7 cells treated with HMGBl (HMG-1), lipopolysaccharide (LPS), or HMGB 1 B box (B box).
FIG. 11B is a histogram ofthe effect of HMGBl (HMG-1) or HMGBl B box (B Box) polypeptide stimulation on activation ofthe NF-κB-dependent ELAM promoter (measured by luciferase activity) in RAW 264.7 cells co-transfected with a murine MyD 88 -dominant negative (+MyD 88 DN) mutant (corresponding to amino acids 146-296), or empty vector (-MyD 88 DN). Data are expressed as the ratio
(fold-activation) of average luciferase values from unstimulated and stimulated cells (subtracted for background) + SD.
FIG. 12A is the amino acid sequence of a human HMG1 polypeptide (SEQ IDNO:l). FIG. 12B is the amino acid sequence of rat and mouse HMG1 (SEQ ID
NO:2). '
FIG. 12C is the amino acid sequence of human HMG2 (SEQ ID NO:3).
FIG. 12D is the amino acid sequence of a human, mouse, and rat HMG1 A box polypeptide (SEQ ID NO:4). FIG. 12E is the amino acid sequence of a human, mouse, and rat HMG1 B box polypeptide (SEQ ID NO:5).
FIG. 12F is the nucleic acid sequence of a forward primer for human HMG1 (SEQ ID NO:6).
FIG. 12G is the nucleic acid sequence of a reverse primer for human HMG1 (SEQ ID NO:7).
FIG. 12H is the nucleic acid sequence of a forward primer for the carboxy terminus mutant of human HMG1 (SEQ ID NO:8).
FIG. 121 is the nucleic acid sequence of a reverse primer for the carboxy terminus mutant of human HMG1 (SEQ ID NO:9). FIG. 12J is the nucleic acid sequence of a forward primer for the amino terminus plus B box mutant of human HMG1 (SEQ ID NO:10). FIG. 12K is the nucleic acid sequence of a reverse primer for the amino terminus plus B box mutant of human HMG1 (SEQ ID NO: 11).
FIG. 12L is the nucleic acid sequence of a forward primer for a B box mutant of human HMG1 (SEQ ID NO: 12). FIG. 12M is the nucleic acid sequence of a reverse primer for a B box mutant of human HMG1 (SEQ ID NO: 13).
FIG. 12N is the nucleic acid sequence of a forward primer for the amino terminus plus A box mutant of human HMG1 (SEQ ID NO: 14).
FIG. 120 is the nucleic acid sequence of a reverse primer for the amino terminus plus A box mutant of human HMG1 (SEQ ID NO:15).
FIG. 13 is a sequence alignment of HMGBl polypeptide sequences from rat (SEQ ID NO:2), mouse (SEQ ID NO:2), and human (SEQ ID NO: 18).
FIG. 14A is the nucleic acid sequence of HMG1L5 (formerly HMG1L10) (SEQ ID NO: 32) encoding an HMGB polypeptide. FIG. 14B is the polypeptide sequence of HMG1L5 (formerly HMG1L10)
(SEQ ID NO: 24) encoding an HMGB polypeptide.
FIG. 14C is the nucleic acid sequence of HMG1L1 (SEQ ID NO: 33) encoding an HMGB polypeptide.
FIG. 14D is the polypeptide sequence of HMG1L1 (SEQ ID NO: 25) encoding an HMGB polypeptide.
FIG. 14E is the nucleic acid sequence of HMG1L4 (SEQ ID NO: 34) encoding an HMGB polypeptide.
FIG. 14F is the polypeptide sequence of HMG1L4 (SEQ ID NO: 26) encoding an HMGB polypeptide. FIG. 14G is the nucleic acid sequence ofthe HMG polypeptide sequence of the BAC clone RPl 1-395A23 (SEQ ID NO: 35).
FIG. 14H is the polypeptide sequence ofthe HMG polypeptide sequence of the BAC clone RPl 1-395A23 (SEQ ID NO: 27) encoding an HMGB polypeptide.
FIG. 141 is the nucleic acid sequence of HMG1L9 (SEQ ID NO: 36) encoding an HMGB polypeptide. FIG. 14J is the polypeptide sequence of HMG1L9 (SEQ ID NO: 28) encoding an HMGB polypeptide.
FIG. 14K is the nucleic acid sequence of LOC122441 (SEQ ID NO: 37) encoding an HMGB polypeptide. FIG. 14L is the polypeptide sequence of LOC122441 (SEQ ID NO: 29) encoding an HMGB polypeptide.
FIG. 14M is the nucleic acid sequence of LOC139603 (SEQ ID NO: 38) encoding an HMGB polypeptide.
FIG. 14N is the polypeptide sequence of LOC139603 (SEQ ID NO: 30) encoding an HMGB polypeptide.
FIG. 14O is the nucleic acid sequence of HMG1L8 (SEQ ID NO: 39) encoding an HMGB polypeptide.
FIG. 14P is the polypeptide sequence of HMG1L8 (SEQ ID NO: 31) encoding an HMGB polypeptide.
DETAILED DESCRIPTION OF THE INVENTION
The practice ofthe present invention will employ, unless otherwise indicated, conventional techniques of cell culture, molecular biology, microbiology, cell biology, and immunology, which are well within the skill ofthe art. Such techniques are fully explained in the literature. See, e.g., Sambrook et al., 1989, "Molecular Cloning: A Laboratory Manual", Cold Spring Harbor Laboratory Press; Ausubel et al. (1995), "Short Protocols in Molecular Biology", John Wiley and Sons; Methods in Enzymology (several volumes); Methods in Cell Biology (several volumes), and Methods in Molecular Biology (several volumes).
The present invention is based on a series of discoveries that further elucidate various characteristics ofthe ability of HMGBl to induce production of proinflammatory cytokines and inflammatory cytokine cascades. Specifically, it has been discovered that the proinflammatory active domain of HMGBl is the B box (and in particular, the first 20 amino acids ofthe B box), and that antibodies specific to the B box will inhibit proinflammatory cytokine release and inflammatory cytokine cascades, with results that can alleviate deleterious symptoms caused by inflammatory cytokine cascades. It has also been discovered that the A box is a weak agonist of inflammatory cytokine release, and competitively inhibits the proinflammatory activity ofthe B box and of HMGBl . It has further been discovered that inhibitors of TNF biological activity can be combined with HMGB A boxes and/or antibodies to HMGB 1 , to form pharmaceutical compositions for use in treating conditions characterized by activation of an inflammatory cytokine cascade in patients.
As used herein, an "HMGB polypeptide" or an "HMGB protein" is a substantially pure, or substantially pure and isolated polypeptide, that has been separated from components that naturally accompany it, or a synthetically or recombinantly produced polypeptide having the same amino acid sequence, and increases inflammation, and/or increases release of a proinflammatory cytokine from a cell, and/or increases the activity ofthe inflammatory cytokine cascade. In one embodiment, the HMGB polypeptide has one ofthe above biological activities. In another embodiment, the HMGB polypeptide has two ofthe above biological activities. In a third embodiment, the HMGB polypeptide has all three ofthe above biological activities.
Preferably, the HMGB polypeptide is a mammalian HMGB polypeptide, for example, a human HMGBl polypeptide. Examples of an HMGB polypeptide include a polypeptide comprising or consisting ofthe sequence of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, or SEQ ID NO: 18. Preferably, the HMGB polypeptide contains a B box DNA binding domain and/or an A box DNA binding domain, and/or an acidic carboxyl tenninus as described herein. Other examples of HMGB polypeptides are described in GenBank Accession Numbers AAA64970, AAB08987, P07155, AAA20508, S29857, P09429, NP_002119, CAA31110, S02826, U00431, X67668, NP_005333, NM_016957, and J04179, the entire teachings of which are incorporated herein by reference. Additional examples of HMGB polypeptides include, but are not limited to mammalian HMG1 ((HMGBl) as described, for example, in GenBank Accession Number U51677), HMG2 ((HMGB 2) as described, for example, in GenBanlc Accession Number M83665), HMG-2A ((HMGB3, HMG-4) as described, for example, in GenBank Accession Numbers NM_005342 andNP_005333), HMG14 (as described, for example, in GenBank Accession Number P05114), HMG17 (as described, for example, in GenBank Accession Number X13546), HMGI (as described, for example, in GenBanlc Accession Number L17131), and HMGY (as described, for example, in GenBank Accession Number M23618); nonmammalian HMG Tl (as described, for example, in GenBank Accession Number X02666) and HMG T2 (as described, for example, in GenBank Accession Number L32859) (rainbow trout); HMG-X (as described, for example, in GenBanlc Accession Number D30765) (Xenopus), HMG D (as described, for example, in GenBank Accession Number X71138) and HMG Z (as described, for example, in GenBank Accession Number X71139) (Drosophila); NHP10 protein (HMG protein homolog NHP 1) (as described, for example, in GenBank Accession Number Z48008) (yeast); non-histone chromosomal protein (as described, for example, in GenBank Accession Number O00479) (yeast); HMG 1/ 2 like protein (as described, for example, in GenBank Accession Number Zl 1540) (wheat, maize, soybean); upstream binding factor (UBF-1) (as described, for example, in GenBank Accession Number X53390); PMS1 protein homolog 1 (as described, for example, in GenBank Accession Number U13695); single-strand recognition protein (SSRP, structure-specific recognition protein) (as described, for example, in GenBank Accession Number M86737); the HMG homolog TDP-1 (as described, for example, in GenBanlc Accession Number M74017); mammalian sex-determining region Y protein (SRY, testis-deteπnining factor) (as described, for example, in GenBanlc Accession Number X53772); fungal proteins: mat-1 (as described, for example, in GenBanlc Accession Number AB009451), ste 11 (as described, for example, in GenBank Accession Number X53431) and Mc 1 ; SOX 14 (as described, for example, in GenBanlc Accession Number AF 107043), as well as SOX 1 (as described, for example, in GenBank Accession Number Y13436), SOX 2 (as described, for example, in GenBanlc Accession Number Z31560), SOX 3 (as described, for example, in GenBanlc Accession Number X71135), SOX 6 (as described, for example, in GenBanlc Accession Number AF309034), SOX 8 (as described, for example, in GenBanlc Accession Number AF226675), SOX 10 (as described, for example, in GenBanlc Accession Number AJ001183), SOX 12 (as described, for example, in GenBank Accession Number X73039) and SOX 21 (as described, for example, in GenBank Accession Number AF 107044)); lymphoid specific factor (LEF-1) (as described, for example, in GenBank Accession Number X58636); T-cell specific transcription factor (TCF-1) (as described, for example, in GenBanlc Accession Number X59869); MTT1 (as described, for example, in GenBank Accession Number M62810); and SP100-HMG nuclear autoantigen (as described, for example, in GenBank Accession Number U36501).
Other examples of HMGB proteins are polypeptides encoded by HMGB nucleic acid sequences having GenBank Accession Numbers NG_000897 (HMG1L5 (formerly HMG1L10)) (and in particular by nucleotides 150-797 of NG_000897, as shown in FIGS. 14A and 14B); AF076674 (HMG1L1) (and in particular by nucleotides 1-633 of AF076674, as shown in FIGS. 14C and 14D; AF076676 (HMG1L4) (and in particular by nucleotides 1-564 of AF076676, as shown in FIGS. 14E and 14F); AC010149 (HMG sequence from BAC clone RPl 1-395A23) (and in particular by nucleotides 75503-76117 of AC010149), as shown in FIGS. 14G and 14H); AF165168 (HMG1L9) (and in particular by nucleotides 729-968 of AF165168, as shown in FIGS. 141 and 14J); XM_063129 (LOC122441) (and in particular by nucleotides 319-558 of XM_063129, as shown in FIGS. 14K and 14L); XM_066789 (LOC139603) (and in particular by nucleotides 1-258 ofXM_066789, as shown in FIGS. 14M and 14N); and AF165167 (HMG1L8) (and in particular by nucleotides 456-666 of AF165167, as shown in FIGS. 14O and 14P).
The HMGB polypeptides ofthe present invention also encompass sequence variants. Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other variants. Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by an HMGB nucleic acid molecule, and complements and portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising the nucleotide sequence of an HMGB nucleic acid molecule. Examples of HMGB nucleic acid molecules are known in the art and can be derived from HMGB polypeptides as described herein. Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods.
Preferably, the HMGB polypeptide has at least 60%, more preferably, at least 70%, 75%, 80%, 85%, or 90%, and most preferably at least 95%, sequence identity to a sequence selected from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3 and SEQ ID NO.T8, as determined using the BLAST program and parameters described herein and one of more of the biological activities of an HMGB polypeptide.
In other embodiments, the present invention is directed to an HMGB polypeptide fragment that has HMGB biological activity. By an "HMGB polypeptide fragment that has HMGB biological activity" or a "biologically active HMGB fragment" is meant a fragment of an HMGB polypeptide that has the activity of an HMGB polypeptide. An example of such an HMGB polypeptide fragment is the HMGB B box, as described herein. Biologically active HMGB fragments can be generated using standard molecular biology techniques and assaying the function of the fragment by determining if the fragment, when administered to a cell, increases release of a proinflammatory cytokine from the cell, compared to a suitable control, for example, using methods described herein.
As used herein, an "HMGB A box", also referred to herein as an "A box", is a substantially pure, or substantially pure and isolated polypeptide, that has been separated from components that naturally accompany it, and consists of an amino acid sequence that is less than a full length HMGB polypeptide and which has one or more ofthe following biological activities: inhibiting inflammation, and/or inhibiting release of a proinflammatory cytokine from a cell, and/or decreasing the activity of the inflammatory cytokine cascade. In one embodiment, the HMGB A box polypeptide has one ofthe above biological activities. In another embodiment, the HMGB A box polypeptide has two ofthe above biological activities. In a third embodiment, the HMGB A box polypeptide has all three ofthe above biological activities. Preferably, the HMGB A box has no more than 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, ofthe biological activity of a full length HMGB polypeptide. In one embodiment, the HMGB A box amino acid consists ofthe sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal.
An HMGB A box is also a recombinantly produced polypeptide having the same amino acid sequence as the A box sequences described above. Preferably, the HMGB A box is a mammalian HMGB A box, for example, a human HMG1 A box. The HMGB A box polypeptides ofthe present invention preferably comprise or consist ofthe sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal. An HMGB A box often has no more than about 85 amino acids and no fewer than about 4 amino acids. Examples of polypeptides having A box sequences within them include, but are not limited to, the HMGB proteins and polypeptides described herein. The A box sequences in such polypeptides can be determined and isolated using methods described herein, for example, by sequence comparisons to A boxes described herein and testing for A box biological activity using methods described herein or other methods known in the art.
Additional examples of HMGB A box polypeptide sequences include the following sequences: PDASVNFSEF SKKCSERWKT MSAKEKGKFE
DMAKADKARY EREMKTYIPP KGET (human HMGBl; SEQ ID NO: 40); DSSVNFAEF SKKCSERWKT MSAKEKSKFE DMAKSDKARY DREMKNYVPP KGDK (human HMGB2; SEQ ID NO: 41); PEVPVNFAEF SKKCSERWKT VSGKEKSKFD EMAKADKVRY DREMKDYGPA KGGK (human HMGB3; SEQ ID NO: 42); PDASVNFSEF SKKCSERWKT MSAKEKGKFE DMAKADKARY EREMKTYIPP KGET (HMG1L5 (formerly HMG1L10); SEQ ID NO: 43); SDASVNFSEF SNKCSERWKT MSAKEKGKFE DMAKADKTHY ERQMKTYIPP KGET (HMG1L1; SEQ ID NO: 44); PDASVNFSEF SKKCSERWKA MSAKDKGKFE DMAKVDKADY EREMKTYIPP KGET (HMG1L4; SEQ ID NO: 45); PDASVKFSEF LKKCSETWKT IFAKEKGKFE DMAKADKAHY EREMKTYIPP KGEK (HMG sequence from BAC clone RPl 1- 395A23; SEQ ID NO: 46); PDASLNFSEF SQKCPETWKT TIAKEKGKFE DMAKADKAHY EREMKTYIPP KGET (HMG1L9; SEQ ID NO: 47); PDASVNSSEF SKKCSERWKTMPTKQGKFE DMAKADRAH (HMG1L8; SEQ ID NO: 48); PDASVNFSEF SKKCLVRGKT MSAKEKGQFE AMARADKARY EREMKTYIP PKGET (LOC122441; SEQ ID NO: 49); LDASVSFSEF SNKCSERWKT MSVKEKGKFE DMAKADKACY EREMKIYPYL KGRQ (LOC139603; SEQ ID NO: 50); and GKGDPKKPRG KMSSYAFFVQ TCREEHKKKH PDASVNFSEF SKKCSERWKT MSAKEKGKFE DMAKADKARY EREMKTYIPP KGET (human HMGBl A box; SEQ ID NO: 57). The HMGB A box polypeptides ofthe present invention also encompass sequence variants. Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other variants. Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by an HMGB A box nucleic acid molecule, and complements and portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising the nucleotide sequence of an HMGB A box nucleic acid molecule. Examples of HMGB A box nucleic acid molecules are known in the art and can be derived from HMGB A polypeptides as described herein. Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods. Preferably, an HMGB A box has at least 60%, more preferably, at least 70%, 75%, 80%, 85%, or 90%, and most preferably at least 95%, sequence identity to an HMGB A box polypeptide described herein, for example, the sequence of SEQ ID NO:4, SEQ ID NO:22, or SEQ ID NO:57, as determined using the BLAST program and parameters described herein, and one of more ofthe biological activities of an HMGB A box, as determined using methods described herein or other method known in the art. The present invention also features A box biologically active fragments. By an "A box fragment that has A box biological activity" or an "A box biologically active fragment" is meant a fragment of an HMGB A box that has the activity of an HMGB A box, as described herein. For example, the A box fragment can decrease release of a pro-inflammatory cytokine from a vertebrate cell, decrease inflammation, and/or decrease activity ofthe inflammatory cytokine cascade. A box fragments can be generated using standard molecular biology techniques and assaying the function ofthe fragment by determining if the fragment, when administered to a cell inhibits release of a proinflammatory cytokine from the cell, for example, using methods described herein. A box biologically active fragments can be used in the methods described herein in which full length A box polypeptides are used, for example, inhibiting release of a proinflammatory cytokine from a cell, or treating a patient having a condition characterized by activation of an inflammatory cytokine cascade. As used herein, an "HMGB B box", also referred to herein as a "B box", is a substantially pure, or substantially pure and isolated polypeptide, that has been separated from components that naturally accompany it, and consists of an amino acid sequence that is less than a full length HMGB polypeptide and has one or more ofthe following biological activities: increasing inflammation, increasing release of a proinflammatory cytokine from a cell, and or increasing the activity ofthe inflammatory cytokine cascade. In one embodiment, the HMGB B box polypeptide has one ofthe above biological activities. In another embodiment, the HMGB B box polypeptide has two ofthe above biological activities. In a third embodiment, the HMGB B box polypeptide has all three ofthe above biological activities. Preferably, the HMGB B box has at least 25%, 30%, 40%, 50%, 60%, 70%, 80% or 90%, ofthe biological activity of a full length HMGB polypeptide. In another embodiment, the HMGB B box does not comprise an HMGB A box.
In another embodiment, the HMGB B box is a polypeptide that is about 90%, 80%, 70%, 60%, 50%, 40%, 35%, 30%, 25%, or 20%, ofthe length of a full length HMGB 1 polypeptide. In another embodiment, the HMGB B box comprises or consists ofthe sequence of SEQ ID NO:5, SEQ ID NO:20 or SEQ ID NO:58, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal, but is still less than the full length HMGB polypeptide. An HMGB B box polypeptide is also a recombinantly produced polypeptide having the same amino acid sequence as an HMGB B box polypeptide described above. Preferably, the HMGB B box is a mammalian HMGB B box, for example, a human HMGBl B box. An HMGB B box often has no more than about 85 amino acids and no fewer than about 4 amino acids. Examples of polypeptides having B box sequences within them include, but are not limited to, the HMGB proteins and polypeptides described herein. The B box sequences in such polypeptides can be determined and isolated using methods described herein, for example, by sequence comparisons to B boxes described herein and testing for biological activity, using methods described herein or other methods known in the art.
Additional examples of HMGB B box polypeptide sequences include the following sequences: FKDPNAPKRP PSAFFLFCSE YRPKIKGEHP GLSIGDNAKK LGEMWΝΝTAA DDKQPYEKKA AKLKEKYEKD IAAY (human HMGB 1 ; SEQ ID NO: 51); KKDPNAPKRP PSAFFLFCSE HRPKIKSEHP GLSIGDTAKK LGEMWSEQSA KDKQPYEQKA AKLKEKYEKD IAAY (human HMGB2; SEQ ID NO: 52); FKDPNAPKRL PSAFFLFCSE YRPKIKGEHP GLSIGDNAKK LGEMWΝΝTAA DDKQPYEKKA AKLKEKYEKD IAAY (HMG1L5 (formerly HMG1L10); SEQ ID NO: 53); FKDPNAPKRP PSAFFLFCSE YHPKIKGEHP GLSIGDNAKK LGEMWΝΝTAA DDKQPGEKKA AKLKEKYEKD IAAY (HMG1L1; SEQ ID NO: 54); FKDSNAPKRP PSAFLLFCSE YCPKIKGEHP GLPISDVAKK LVEMWNNTFA DDKQLCEKKA AKLKEKYKKD TATY (HMG1L4; SEQ ID NO: 55); FKDPNAPKRP PSAFFLFCSE YRPKIKGEHP GLSIGD KK LAGMWNNTAA ADKQFYEKKA AKLKEKYKKD IAAY (HMG sequence from BAC clone RP 11 -359A23 ; SEQ ID NO: 56); and FKDPNAPKRP PSAFFLFCSE YRPKIKGEHP GLSIGDNAKK LGEMWΝΝTAA DDKQPYEKKA AKLKEKYEKD IAAYRAKGKP DAAKKGNNKA EK (human HMGBl box; SEQ ID NO: 58).
The HMGB B box polypeptides ofthe invention also encompass sequence variants. Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other variants. Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by an HMGB box nucleic acid molecule, and complements and portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising the nucleotide sequence of an HMGB B box nucleic acid molecule. Examples of HMGB B box nucleic acid molecules are known in the art and can be derived from HMGB B box polypeptides as described herein. Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods. Preferably, a non-naturally occurring HMGB B box polypeptide has at least 60%, more preferably, at least 70%, 75%, 80%, 85%, or 90%, and most preferably at least 95%, sequence identity to the sequence of an HMGB B box as described herein, for example, the sequence of SEQ ID NO:5, SEQ ID NO:20, or SEQ ID NO:58, as determined using the BLAST program and parameters described herein. Preferably, the HMGB B box consists ofthe sequence of SEQ ID NO:5, SEQ ID NO:20, or SEQ ID NO:58, or the amino acid sequence in the corresponding region of an HMGB protein in a mammal, and has one or more ofthe biological activities of an HMGB B box, as detennined using methods described herein or other methods known in the art.
In other embodiments, the present invention is directed to a polypeptide comprising an HMGB B box biologically active fragment that has B box biological activity, or a non-naturally occurring HMGB B box fragment In another embodiment, the present invention is directed to a polypeptide comprising a vertebrate HMGB B box or a fragment thereof that has B box biological activity, or a non-naturally occurring HMGB B box but not comprising a full length HMGB polypeptide. By a "B box fragment that has B box biological activity" or a "B box biologically active fragment" is meant a fragment of an HMGB B box that has the activity of an HMGB B box. For example, the B box fragment can induce release of a pro-inflammatory cytokine from a vertebrate cell or increase inflammation, or induce the inflammatory cytokine cascade. An example of such a B box fragment is the fragment comprising the first 20 amino acids ofthe HMGBl B box (SEQ ID NO: 16 or SEQ ID NO:23), as described herein. B box fragments can be generated using standard molecular biology techniques and assaying the function ofthe fragment by determining if the fragment, when administered to a cell, increases release of a proinflammatory cytokine from the cell, as compared to a suitable control, for example, using methods described herein or other methods known in the art. HMGB polypeptides, HMGB A boxes, and HMGB B boxes, either naturally occurring or non-naturally occurring, include polypeptides that have sequence identity to the HMGB polypeptides, HMGB A boxes, and HMGB B boxes described herein. As used herein, two polypeptides (or a region ofthe polypeptides) are substantially homologous or identical when the amino acid sequences are at least about 60%, 70%, 75%, 80%, 85%, 90%, or 95% or more, homologous or identical. The percent identity of two amino acid sequences (or two nucleic acid sequences) can be determined by aligning the sequences for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first sequence). The amino acids or nucleotides at corresponding positions are then compared, and the percent identity between the two sequences is a function ofthe number of identical positions shared by the sequences (i.e., % identity = # of identical positions/total # of positions x 100). In certain embodiments, the length ofthe HMGB polypeptide, HMGB A box polypeptide, or HMGB B box polypeptide aligned for comparison purposes is at least 30%), preferably, at least 40%, more preferably, at least 60%, and even more preferably, at least 70%>, 80%>, 90%, or 100%, ofthe length ofthe reference sequence, for example, those sequence provided in FIGS. 12A-12E, FIGS. 14A-14P, and SEQ ID NOS: 18, 20, and 22. The actual comparison ofthe two sequences can be accomplished by well-known methods, for example, using a mathematical algorithm. A preferred, non-limiting example of such a mathematical algorithm is described in Karlin et al. (Proc. Natl. Acad. Sci. USA, 90:5873-5877, 1993). Such an algorithm is incorporated into the BLASTN and BLASTX programs (version 2.2) as described in Schaffer et al. (Nucleic Acids Res., 29:2994-3005, 2001). When utilizing BLAST and Gapped BLAST programs, the default parameters ofthe respective programs (e.g. , BLASTN) can be used. See the Internet site for the National Center for Biotechnology Information (NCBI). In one embodiment, the database searched is a non-redundant (NR) database, and parameters for sequence comparison can be set at: no filters; Expect value of 10; Word Size of 3; the Matrix is BLOSUM62; and Gap Costs have an Existence of 11 and an Extension of 1.
Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, CABIOS (1989). Such an algorithm is incorporated into the ALIGN program (version 2.0), which is part ofthe GCG (Accelrys) sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12 , and a gap penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art and include ADVANCE and ADAM as described in Torellis and Robotti (Comput. Appl. Biosci., 10: 3-5,1994); and FASTA described in Pearson and Lipman (Proc. Natl. Acad. Sci USA, 85: 2444-2448, 1988).
In another embodiment, the percent identity between two amino acid sequences can be accomplished using the GAP program in the GCG software package (Accelrys, San Diego, California) using either a Blossom 63 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4. In yet another embodiment, the percent identity between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package (Accelrys, San Diego, California), using a gap weight of 50 and a length weight of 3.
As used herein, a "cytokine" is a soluble protein or peptide which is naturally produced by mammalian cells and which acts in vivo as a humoral regulator at micro- to picomolar concentrations. Cytokines can, either under normal or pathological conditions, modulate the functional activities of individual cells and tissues. A proinflammatory cytokine is a cytokine that is capable of causing any ofthe following physiological reactions associated with inflammation: vasodilation, hyperemia, increased permeability of vessels with associated edema, accumulation of granulocytes and mononuclear phagocytes, or deposition of fibrin. In some cases, the proinflammatory cytokine can also cause apoptosis, such as in chronic heart failure, where TNF has been shown to stimulate cardiomyocyte apoptosis (Pulkki, Ann. Med. 29: 339-343, 1997; and Tsutsui et al, Immunol. Rev. 174:192-209, 2000). Nonlimiting examples of proinflammatory cytokines are tumor necrosis factor (TNF), interleulcin (IL)-l , IL-lβ, IL-6, IL-8, IL-18, interferon γ, HMG-1, platelet-activating factor (PAF), and macrophage migration inhibitory factor (MIF). Proinflammatory cytokines are to be distinguished from anti-inflammatory cytokines, such as IL-4, IL-10, and IL-13, which are not mediators of inflammation. In many instances, proinflammatory cytokines are produced in an inflammatory cytokine cascade, defined herein as an in vivo release of at least one proinflammatory cytokine in a mammal, wherein the cytokine release affects a physiological condition ofthe mammal. Thus, an inflammatory cytokine cascade is inhibited in embodiments of the invention where proinflammatory cytokine release causes a deleterious physiological condition.
As used herein, "an agent that inhibits TNF biological activity" is an agent that decreases one or more ofthe biological activities of TNF. Examples of TNF biological activity include, but are not limited to, vasodilation, hyperemia, increased permeability of vessels with associated edema, accumulation of granulocytes and mononuclear phagocytes, and deposition of fibrin. Agents that inhibit TNF biological activity include agents that inhibit (decrease) the interaction between TNF and a TNF receptor. Examples of such agents include antibodies or antigen binding fragments thereof that bind to TNF, antibodies or antigen binding fragments that bind a TNF receptor, and molecules that bind TNF or the TNF receptor and prevent
TNF/TNF receptor interaction. Such agents include, but are not limited to peptides, proteins, synthesized molecules, for example, synthetic organic molecules, naturally-occurring molecule, for example, naturally occurring organic molecules, nucleic acid molecules, and components thereof. Preferred examples of agents that inhibit TNF biological activity include infliximab (Remicade; Centocor, Inc.,
Malvern, Pennsylvania), etanercept (Immunex; Seattle, Washington), adalimumab (D2E7; Abbot Laboratories, Abbot Park Illinois), CDP870 (Pharmacia Corporation; Bridgewater, New Jersey) CDP571 (Celltech Group pic, United Kingdom), Lenercept (Roche, Switzerland), and Thalidomide.
Inflammatory cytokine cascades contribute to deleterious characteristics, including inflammation and apoptosis, of numerous disorders. Included are disorders characterized by both localized and systemic reactions, including, without limitation, the disorders described herein (e.g., those conditions enumerated in the background section of this specification). Particular disorders characterized by inflammatory cytokine cascades include, e.g., sepsis, allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstructive pulmonary disease, psoriasis, pancreatitis, peritonitis, burns, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative colitis, multiple sclerosis, and cachexia.
A Box Polypeptides and Biologically Active Fragments Thereof As described herein, in one aspect the present invention is directed to a polypeptide composition comprising a vertebrate HMGB A box, or a biologically active fragment thereof, which can inhibit release of a piOinflammatory cytokine from a cell treated with HMG, or which can be used to treat a condition characterized by activation of an inflammatory cytokine cascade. In certain embodiments, the invention is directed to compositions comprising an HMGB A box, or a biologically active fragment or variant thereof, in combination with one or more agents that inhibit TNF biological activity, for example, infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, or Thalidomide. Such compositions can be used to inhibit release of a proinflammatory cytokine from a vertebrate cell treated with HMG, and/or can be used to treat a condition characterized by activation of an inflammatory cytokine cascade.
When referring to the effect of any ofthe compositions or methods ofthe invention on the release of proinflammatory cytokines, the use ofthe terms "inhibit" or "decrease" encompasses at least a small but measurable reduction in proinflammatory cytokine release. In preferred embodiments, the release ofthe proinflammatory cytokine is inhibited by at least 20% over non-treated controls; in more preferred embodiments, the inhibition is at least 50%; in still more preferred embodiments, the inhibition is at least 70%, and in the most preferred embodiments, the inhibition is at least 80%. Inhibition can be assessed using methods described herein or other methods known in the art. Such reductions in proinflammatory cytokine release are capable of reducing the deleterious effects of an inflammatory cytokine cascade in in vivo embodiments.
Because HMGB A boxes (e.g., vertebrate HMGB A boxes) show a high degree of sequence conservation (see, for example, FIG. 13 for an amino acid sequence comparison of rat, mouse, and human HMGB polypeptides), it is believed that an HMGB A box (e.g., a vertebrate HMGB A box) can inhibit release of a proinflammatory cytokine from a vertebrate cell treated with HMGB. Therefore, an HMGB A box (e.g., a vertebrate HMGB A box) is within the scope ofthe invention. Preferably, the HMGB A box is a vertebrate HMGB A box (e.g., a mammalian HMGB A box, such as a human HMGB 1 A box provided herein as SEQ ID NO :4, SEQ ID NO:22, or SEQ ID NO:57). Also included in the present invention are fragments ofthe HMGBl A box having HMGB A box biological activity, as described herein.
It would also be recognized by the skilled artisan that non-naturally occurring HMGB A boxes (or biologically active fragments thereof) can be created without undue experimentation, which would inhibit release of a proinflammatory cytokine from a vertebrate cell treated with a vertebrate HMGB. These non-naturally occurring functional A boxes (variants) can be created by aligning amino acid sequences of HMGB A boxes from different sources, and making one or more substitutions in one ofthe sequences at amino acid positions where the A boxes differ. The substitutions are preferably made using the same amino acid residue that occurs in the compared A box. Alternatively, a conservative substitution is made from either ofthe residues.
Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains. Conservatively substituted amino acids can be grouped according to the chemical properties of their side chains. For example, one grouping of amino acids includes those amino acids have neutral and hydrophobic side chains (a, v, 1, i, p, w, f, and m); another grouping is those amino acids having neutral and polar side chains (g, s, t, y, c, n, and q); another grouping is those amino acids having basic side chains (k, r, and h); another grouping is those amino acids having acidic side chains (d and e); another grouping is those amino acids having aliphatic side chains (g, a, v, 1, and i); another grouping is those amino acids having aliphatic-hydroxyl side chains (s and t); another grouping is those amino acids having amine-containing side chains (n, q, k, r, and h); another grouping is those amino acids having aromatic side chains (f, y, and w); and another grouping is those amino acids having sulfur-containing side chains (c and m). Preferred conservative amino acid substitutions groups are: r-k; e-d, y-f, 1-m; v-i, and q-h.
While a conservative amino acid substitution would be expected to preserve the biological activity of an HMGB A box polypeptide, the following is one example of how non-naturally occurring A box polypeptides (variants) can be made by comparing the human HMGB 1 A box (SEQ ID NO :4) with residues 32 to 85 of SEQ ID NO:3 ofthe human HMGB2 A box (SEQ ID NO:17).
HMGBl pdasvnfsef skkcserwkt msakekgkfe dmakadkary eremktyipp kget (SEQ ID NO:4)
HMGB2 pdssvnfaef skkcserwkt msakekskfe dmaksdkary dremknyvpp kgdk (SEQ ID NO: 17)
A non-naturally occurring HMGB A box can be created by, for example, by substituting the alanine (a) residue at the third position in the HMGBl A box with the serine (s) residue that occurs at the third position ofthe HMGB2 A box. The skilled artisan would know that the substitution would provide a functional non- naturally occurring A box because the s residue functions at that position in the HMGB2 A box. Alternatively, the third position ofthe HMGBl A box can be substituted with any amino acid that is conservative to alanine or serine, such as glycine (g), threonine (t), valine (v) or leucine (1). The skilled artisan would recognize that these conservative substitutions would be expected to result in a functional A box because A boxes are not invariant at the third position, so a conservative substitution would provide an adequate structural substitute for an amino acid that is naturally occurring at that position.
Following the above method, a great many non-naturally occurring HMGB A boxes could be created without undue experimentation wliich would be expected to be functional, and the functionality of any particular non-naturally occurring HMGB A box could be predicted with adequate accuracy. In any event, the functionality of any non-naturally occurring HMGB A box could be determined without undue experimentation by simply adding it to cells along with an HMGB polypeptide, and determining whether the A box inhibits release of a proinflammatory cytokine by the cells, using, for example, methods described herein.
The cell from which the A box or an A box biologically active fragment will inhibit the release of HMG-induced proinflammatory cytokines can be any cell that can be induced to produce a proinflammatory cytokine. In preferred embodiments, the cell is a mammalian cell, for example, an immune cell (e.g., a macrophage, a monocyte, or a neutrophil).
Polypeptides comprising an A box or A box biologically active fragment that can inhibit the production of any single proinflammatory cytokine, now known or later discovered, are within the scope ofthe present invention. Preferably, the antibodies can inhibit the production of TNF, IL- 1 β, and/or IL-6. Most preferably, the antibodies can inhibit the production of any proinflammatory cytokines produced by the vertebrate cell.
B Box Polypeptides and Biologically Active Fragments Thereof
As described herein, in one aspect the present invention is directed to a polypeptide composition comprising a vertebrate HMGB B box, or a biologically active fragment thereof, which can increase release of a proinflammatory cytokine from a vertebrate cell treated with HMGB.
When referring to the effect of any ofthe compositions or methods ofthe invention on the release of proinflammatory cytokines, the use ofthe term "increase" encompasses at least a small but measurable rise in proinflammatory cytokine release. In preferred embodiments, the release ofthe proinflammatory cytokine is increased by at least 1.5-fold, at least 2-fold, at least 5-fold, or at least 10-fold, over non-treated controls. Such increases in proinflammatory cytokine release are capable of increasing the effects of an inflammatory cytokine cascade in in vivo embodiments. Such polypeptides can also be used to induce weight loss and/or treat obesity.
Because all HMGB B boxes show a high degree of sequence conservation (see, for example, FIG. 13 for an amino acids sequence comparison of rat, mouse, and human HMGB polypeptides), it is believed that functional non-naturally occurring HMGB B boxes can be created without undue experimentation by making one or more conservative amino acid substitutions, or by comparing naturally occurring vertebrate B boxes from different sources and substituting analogous amino acids, as was discussed above with respect to the creation of functional non- naturally occurring A boxes. In particularly preferred embodiments, the B box comprises SEQ ID NO:5, SEQ ID NO: 20 or SEQ ID NO:58, which are the sequences (three different lengths) ofthe human HMGBl B box, or, comprises the B box sequences from the polypeptides shown in FIGS. 14A-14P, or is a fragment of an HMGB B box that has B box biological activity. For example, a 20 amino acid sequence contained within SEQ ID NO: 20 contributes to the function ofthe B box. This 20 amino acid B-box fragment has the following amino acid sequence: flcdpnapkrl psafflfcse (SEQ ID NO:23). Another example of an HMGB B box biologically active fragment consists of amino acids 1-20 of SEQ ID NO: 5 (naplcrppsafflfcseyrpk; SEQ ID NO: 16).
Antibodies to HMGB and HMGB B Box Polypeptides The invention is also directed to a purified preparation of antibodies that bind to an HMGB polypeptide or a biologically active fragment thereof (anti-HMGB antibodies). The anti-HMGB antibodies can be neutralizing antibodies (i.e., can inhibit a biological activity of an HMG polypeptide or a biologically active fragment thereof, for example, the release of a proinflammatory cytokine from a vertebrate cell induced by HMG). The invention is also directed to a purified preparation of antibodies that specifically bind to a vertebrate high mobility group protein (HMG) B box or a biologically active fragment thereof, but do not selectively bind to non-B box epitopes of HMGB (anti-HMGB B box antibodies). In these embodiments, the antibodies can also be neutralizing antibodies (i.e., they can inhibit a biological activity of a B box polypeptide or biologically active fragment thereof, for example, the release of a proinflammatory cytokine from a vertebrate cell induced by HMGB). Such antibodies can be combined with one or more agents that inhibit TNF biological activity, for example, infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, or Thalidomide. The term "antibody" or "purified antibody" as used herein refers to immunoglobulm molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that selectively binds an antigen. A molecule that selectively binds to a polypeptide ofthe invention is a molecule that binds to that polypeptide or a fragment thereof, but does not substantially bind other molecules in a sample, e.g., a biological sample that naturally contains the polypeptide. Preferably the antibody is at least 60%, by weight, free from proteins and naturally occurring organic molecules with which it is naturally associated. More preferably, the antibody preparation is at least 75% or 90%, and most preferably, 99%, by weight, antibody. Examples of immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments that can be generated by treating the antibody with an enzyme such as pepsin.
The invention provides polyclonal and monoclonal antibodies that selectively bind to a HMGB B box polypeptide ofthe invention. The term "monoclonal antibody" or "monoclonal antibody composition," as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of a polypeptide ofthe invention. A monoclonal antibody composition thus typically displays a single binding affinity for a particular polypeptide ofthe invention with which it immunoreacts. Polyclonal antibodies can be prepared, e.g., as described herein, by immunizing a suitable subject with a desired immunogen, e.g., an HMGB B box polypeptide ofthe invention or fragment thereof. The antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide. If desired, the antibody molecules directed against the polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction.
At an appropriate time after immunization, e.g., when the antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (Nature 256:495-497, 1975), the human B cell hybridoma technique (Kozbor et al., Immunol. Today 4:72, 1983), the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96, 1985) or trioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology, Coligan et al, (eds.) John Wiley & Sons, Inc., New York, NY, 1994). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an immunogen as described above, and the culture supernatants ofthe resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds a particular polypeptide (e.g., a polypeptide ofthe invention).
Any ofthe many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating a monoclonal antibody to a polypeptide ofthe invention (see, e.g., Current Protocols in Immunology, supra; Galfre et al. (Nature, 266:55052, 1977); R.H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses, Plenum Publishing Corp., New York, New York (1980); and Lerner (Yale J. Biol. Med. 54:387-402, 1981)). Moreover, the ordinarily skilled worker will appreciate that there are many variations of such methods that also would be useful.
In one alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody to an HMGB B box polypeptide ofthe invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide to thereby isolate immunoglobulin library members that bind the polypeptide. Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP™ Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Patent No. 5,223,409; PCT Publication No. WO 92/18619; PCT Publication No. WO 91/17271; PCT Publication No. WO 92/20791; PCT Publication No. WO 92/15679; PCT Publication No. WO 93/01288; PCT Publication No. WO 92/01047; PCT
Publication No. WO 92/09690; PCT Publication No. WO 90/02809; Fuchs et al, Bio/Technology 9:1370-1372, 1991; Hay et al, Hum. Antibod. Hybridomas 3:81-85, 1992; Huse et al. (Science 246:1275-1281, 1989); and Griffiths et al. (EMBO J. 12:725-734, 1993). Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope ofthe invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art. In general, antibodies ofthe invention (e.g., a monoclonal antibody) can be used to isolate an HMGB B box polypeptide ofthe invention by standard techniques, such as affinity chromatography or immunoprecipitation. A polypeptide-specific antibody can facilitate the purification of natural polypeptide from cells and of recombinantly produced polypeptide expressed in host cells. Moreover, an antibody specific for an HMGB B box polypeptide ofthe invention can be used to detect the polypeptide (e.g., in a cellular lysate, cell supernatant, or tissue sample) in order to evaluate the abundance and pattern of expression ofthe polypeptide.
Because vertebrate HMGB polypeptides and HMGB B boxes show a high degree of sequence conservation, it is believed that vertebrate HMGB polypeptides or HMGB B boxes in general can induce release of a proinflammatory cytokine from a vertebrate cell. Therefore, antibodies against vertebrate HMGB polypeptides or HMGB B boxes are within the scope ofthe invention. In one embodiment, the antibodies are neutralizing antibodies.
Preferably, the HMGB polypeptide is a mammalian HMG, as described herein, more preferably a mammalian HMGBl polypeptide, most preferably a human HMGB 1 polypeptide, provided herein as SEQ ID NO: 1. Antibodies can also be directed against an HMGB polypeptide fragment that has HMGB polypeptide biological activity.
Preferably, the HMGB B box is a mammalian HMGB B box, more preferably a mammalian HMGBl B box, most preferably a human HMGBl B box, provided herein as SEQ ID NO:5, SEQ ID NO:20, or SEQ ID NO:58. Antibodies can also be directed against an HMGB B box fragment that has B box biological activity.
Antibodies generated against an HMGB immunogen or an HMGB B box immunogen can be obtained by administering an HMGB polypeptide, or fragment thereof, an HMGB B box or fragment thereof, or cells comprising the HMGB polypeptide, the HMGB B box, or fragments thereof, to an animal, preferably a nonhuman, using routine protocols. The polypeptide, such as an antigenically or immunologically equivalent derivative, is used as an antigen to immunize a mouse or other animal, such as a rat or chicken. The immunogen may be associated, for example, by conjugation, with an immunogenic carrier protein, for example, bovine serum albumin (BSA) or keyhole limpet haemocyanin (KLH). Alternatively, a multiple antigenic peptide comprising multiple copies ofthe HMGB or HMGB B box or fragment, may be sufficiently antigenic to improve immunogenicity so as to obviate the need for a carrier. Bispecific antibodies, having two antigen binding domains where each is directed against a different HMGB or HMGB B box epitope, may also be produced by routine methods.
For preparation of monoclonal antibodies, any technique known in the art that provides antibodies produced by continuous cell line cultures can be used. See, e.g., Kohler and Milstein, supra; and Cole et al, supra.
Techniques for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce single chain antibodies to HMGB, the B box or fragments thereof. Also, transgenic mice, or other organisms such as other mammals, may be used to express humanized antibodies.
If the antibody is used therapeutically in in vivo applications, the antibody is preferably modified to make it less immunogenic in the individual. For example, if the individual is human the antibody is preferably "humanized"; where the complementarity determining region(s) ofthe antibody is transplanted into a human antibody (for example, as described in Jones et al. (Nature 321:522-525, 1986); and Tempest et al. (Biotechnology 9:266-273, 1991)).
Phage display technology can also be utilized to select antibody genes with binding activities towards the polypeptide either from repertoires of PCR amplified v-genes of lymphocytes from humans screened for possessing anti-B box antibodies or from naive libraries (McCafferty et al., Nature 348:552-554, 1990; and Marks, et al., Biotechnology 10:779-783, 1992). The affinity of these antibodies can also be improved by chain shuffling (Clackson et al., Nature 352: 624-628, 1991). When the antibodies are obtained that specifically bind to HMGB epitopes or to HMGB B box epitopes, they can then be screened, without undue experimentation, for the ability to inhibit release of a proinflammatory cytokine. Anti-HMGB B box antibodies that can inhibit the production of any single proinflammatory cytokine, and/or inhibit the release of a proinflammatory cytokine from a cell, and/or inhibit a condition characterized by activation of an inflammatory cytokine cascade, are within the scope ofthe present invention. Preferably, the antibodies can inhibit the production of TNF, IL-1 β, and/or IL-6. Most preferably, the antibodies can inhibit the production of any proinflammatory cytokines produced by the vertebrate cell. For methods of inhibiting release of a proinflammatory cytokine from a cell or treating a condition characterized by activation of an inflammatory cytokine cascade using antibodies to the HMGB B box or a biologically active fragment thereof, the cell can be any cell that can be induced to produce a proinflammatory cytokine. In preferred embodiments, the cell is an immune cell, for example, macrophages, monocytes, or neutrophils. Compositions Comprising One or More of an HMGB A box polypeptide, an Antibody to HMGB, an Antibody to an HMGB B box, and an Inhibitor of TNF Biological Activity
In certain embodiments, the present invention is directed to a composition comprising any ofthe above-described polypeptides (e.g., an HMGB A box polypeptide or biologically active fragment as described herein) in a pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include those described herein. In these embodiments, the composition can inhibit a condition characterized by activation of an inflammatory cytokine cascade. The condition can be one where the inflammatory cytokine cascade causes a systemic reaction, such as with endotoxic shock. Alternatively, the condition can be mediated by a localized inflammatory cytokine cascade, as in rheumatoid arthritis. Nonlimiting examples of conditions which can be usefully treated using the present invention include those conditions enumerated in the background section of this specification. In one embodiment, the condition to be treated is appendicitis, peptic, gastric or duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute or ischemic colitis, hepatitis, Crohn's disease, asthma, allergy, anaphylactic shock, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, septic abortion, disseminated bacteremia, burns, Alzheimer's disease, coeliac disease, congestive heart failure, adult respiratory distress syndrome, cerebral infarction, cerebral embolism, spinal cord injury, paralysis, allograft rejection or graft- versus-host disease, hi another embodiment, the condition is endotoxic shock or allograft rejection. Where the condition is allograft rejection, the composition may advantageously also include an immunosuppressant that is used to inhibit allograft rejection, such as cyclosporin. hi other embodiments, the invention is directed to a composition comprising the antibody preparations described above (e.g., anti-HMGB B box antibodies or biologically active fragments thereof, as described herein), in a pharmaceutically acceptable carrier, hi these embodiments, the compositions can inhibit a condition characterized by the activation of an inflammatory cytokine cascade. Conditions that can be treated with these compositions have been previously enumerated. In other embodiments, the invention is directed to a composition comprising any ofthe above-described HMGB A box polypeptides, and/or an antibody or antigen binding fragment thereof that binds HMGB, and/or an antibody or antigen binding fragment thereof that binds an HMGB B box, and an agent that inhibits TNF biological activity (collectively termed "combination therapy compositions").
Preferred examples of agents that inhibit TNF biological activity include infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide. Such combination therapy compositions can further comprise a pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include those • described herein. In these embodiments, the combination therapy composition can inhibit a condition characterized by activation of an inflammatory cytokine cascade and/or inhibit release of a proinflammatory cytokine from a cell. The condition can be one where the inflammatory cytokine cascade causes a systemic reaction, such as with endotoxic shock. Alternatively, the condition can be mediated by a localized inflammatory cytokine cascade, as in rheumatoid arthritis. Nonlimiting examples of conditions which can be usefully treated using the present invention include those conditions enumerated in the background section of this specification. In one embodiment, the condition to be treated is sepsis, allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstructive pulmonary disease, psoriasis, pancreatitis, peritonitis, burns, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative colitis, multiple sclerosis, and cachexia. Preferably the combination therapy compositions are administered to a patient in need thereof in an amount sufficient to inhibit release of proinflammatory cytokine from a cell and/or to treat a condition characterized by activation of an inflammatory cytokine cascade. In one embodiment, release ofthe proinflammatory cytokine is inhibited by at least 10%, 20%, 25%, 50%, 75%, 80%, 90% or 95%, as assessed using methods described herein or other methods known in the art.
The carrier or excipient included with the polypeptide (e.g., an HMGB A box polypeptide or biologically active fragment thereof), antibody (e.g., an anti-HMGB B box antibody or biologically active fragment thereof) or combination therapy composition (e.g., an HMGB A box polypeptide or biologically active fragment thereof and an agent that inhibits TNF biological activity, and/or an antibody or antigen binding fragment thereof that binds HMGB and an agent that inhibits TNF biological activity, and/or an antibody or antigen binding fragment thereof that binds an HMGB B box and an agent that inhibits TNF biological activity) is chosen based on the expected route of administration ofthe composition in therapeutic applications. The route of administration ofthe composition depends on the condition to be treated. For example, intravenous injection may be preferred for treatment of a systemic disorder such as endotoxic shock, and oral administration may be preferred to treat a gastrointestinal disorder such as a gastric ulcer. The route of administration and the dosage ofthe composition to be administered can be determined by the skilled artisan, without undue experimentation, in conjunction with standard dose-response studies. Relevant circumstances to be considered in making such determinations include the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response ofthe individual patient, and the severity ofthe patient's symptoms. Thus, depending on the condition, the antibody composition can be administered orally, parenterally, intranasally, vaginally, rectally, lingually, sublingually, bucally, intrabuccaly and transdermally to the patient. Accordingly, compositions designed for oral, lingual, sublingual, buccal and intrabuccal administration can be made without undue experimentation by means well known in the art, for example, with an inert diluent or with an edible carrier. The compositions may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the pharmaceutical compositions ofthe present invention may be incorporated with excipients and used in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, chewing gums and the like.
Tablets, pills, capsules, troches and the like may also contain binders, recipients, disintegrating agent, lubricants, sweetening agents, and flavoring agents. Some examples of binders include microcrystalline cellulose, gum tragacanth and gelatin. Examples of excipients include starch and lactose. Some examples of disintegrating agents include alginic acid, corn starch and the like. Examples of lubricants include magnesium stearate and potassium stearate. An example of a glidant is colloidal silicon dioxide. Some examples of sweetening agents include sucrose, saccharin and the like. Examples of flavoring agents include peppermint, methyl salicylate, orange flavoring and the like. Materials used in preparing these various compositions should be pharmaceutically pure and non-toxic in the amounts used.
The compositions ofthe present invention can easily be administered parenterally such as, for example, by intravenous, intramuscular, intrathecal or subcutaneous injection. Parenteral administration can be accomplished by incorporating the compositions ofthe present invention into a solution or suspension. Such solutions or suspensions may also include sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol and/or other synthetic solvents. Parenteral formulations may also include antibacterial agents such as, for example, benzyl alcohol and/or methyl parabens, antioxidants such as, for example, ascorbic acid and/or sodium bisulfite and chelating agents such as EDTA. Buffers, such as acetates, citrates and/or phosphates, and agents for the adjustment of tonicity, such as sodium chloride and/or dextrose, may also be added. The parenteral preparation can be enclosed in ampules, disposable syringes or multiple dose vials made of glass or plastic.
Rectal administration includes administering the pharmaceutical compositions into the rectum or large intestine. This can be accomplished using suppositories or enemas. Suppository formulations can easily be made by methods known in the art. For example, suppository formulations can be prepared by heating glycerin to about 120°C, dissolving the polypeptide composition, antibody composition and/or combination therapy composition in the glycerin, mixing the heated glycerin after which purified water may be added, and pouring the hot mixture into a suppository mold.
Transdermal administration includes percutaneous absorption ofthe composition through the skin. Transdermal formulations include patches, ointments, creams, gels, salves and the like. The present invention includes nasally administering to a mammal (e.g., a human) a therapeutically effective amount ofthe composition. As used herein, nasally administering or nasal administration includes administering the composition to the mucous membranes ofthe nasal passage or nasal cavity ofthe patient. As used herein, pharmaceutical compositions for nasal administration of a composition include therapeutically effective amounts ofthe polypeptide, antibody and/or combination therapy agents, prepared by well-known methods to be administered, for example, as a nasal spray, nasal drop, suspension, gel, ointment, cream or powder. Administration ofthe composition may also take place using a nasal tampon or nasal sponge.
The pharmaceutical compositions (e.g., polypeptide compositions, antibody compositions and/or combination therapy composition) described herein can also include an antagonist of an early sepsis mediator. As used herein, an early sepsis mediator is a proinflammatory cytokine that is released from cells soon (i.e., within 30-60 min.) after induction of an inflammatoiy cytokine cascade (e.g., exposure to LPS). Nonlimiting examples of these cytokines are TNF, IL-l , IL-lβ, IL-6, PAF, and MIF. Also included as early sepsis mediators are receptors for these cytokines (for example, tumor necrosis factor receptor type 1) and enzymes required for production of these cytokines, for example, interleukin-lβ converting enzyme). Antagonists of any early sepsis mediator, now known or later discovered, can be useful for these embodiments by further inhibiting an inflammatory cytokine cascade.
Nonlimiting examples of antagonists of early sepsis mediators are antisense compounds that bind to the mRNA ofthe early sepsis mediator, preventing its expression (see, e.g., Ojwang et al. (Biochemistry 36:6033-6045, 1997); Pampfer et al. (Biol. Reprod. 52:1316-1326, 1995); U.S. Patent No. 6,228,642; Yahata et al. (Antisense Nucleic Acid Drug Dev. 6:55-61, 1996); and Taylor et al. (Antisense Nucleic Acid Drug Dev. 8:199-205, 1998)), ribozymes that specifically cleave the mRNA ofthe early sepsis mediator (see, e.g., Leavitt et al. (Antisense Nucleic Acid Drug Dev. 10: 409-414, 2000); Hendrix et al. (Biochem. J. 314 (Pt. 2): 655-661, 1996)), and antibodies that bind to the early sepsis mediator and inhibit their action (see, e.g., Kam and Targan (Expert Opin. Pharmacother. 1: 615-622, 2000); Nagahira et al. (J. Immunol. Methods 222, 83-92, 1999); Lavine et al. (J. Cereb. Blood Flow Metab. 18: 52-58, 1998); and Holmes et al. (Hybridoma 19: 363-367, 2000)). Any antagonist of an early sepsis mediator, now known or later discovered, is envisioned as within the scope ofthe invention. The skilled artisan can determine the amount of early sepsis mediator to use in these compositions for inhibiting any particular inflammatory cytokine cascade without undue experimentation with routine dose- response studies.
Other agents that can be administered with the compositions described herein include, e.g., Vitaxin™ and other antibodies targeting vβ3 integrin (see, e.g., U.S. Patent No. 5,753,230, PCT Publication Nos. WO 00/78815 and WO 02/070007; the entire teachings of all of which are incorporated herein by reference) and anti-IL-9 antibodies (see, e.g., PCT Publication No. WO 97/08321; the entire teachings of which are incorporated herein by reference). Additional agents that can be administered with the polypeptide compositions described herein include, e.g., B7 antagonists (e.g., CTLA4Ig, anti-CD80 antibodies, anti-CD86 antibodies), methotrexate, and/or CD40 antagonists (e.g., anti-CD40 ligand (CD40L)) (see, e.g., Saito et al., J. Immunol. 160(9):4225-31 (1998)). hi further embodiments, the present invention is also directed to a method of inhibiting the release of a proinflammatory cytokine from a mammalian cell. The method comprises treating the cell with any ofthe HMGB A box compositions, and/or any ofthe HMGB B box or HMGB B box biologically active fragment antibody compositions, and/or any ofthe combination therapy compositions discussed above. It is believed that this method would be useful for inhibiting the cytokine release from any mammalian cell that produces a proinflammatory cytokine. However, in preferred embodiments, the cell is a macrophage, because macrophage production of proinflammatory cytokines is associated with several important diseases.
It is believed that this method is useful for the inhibition of any proinflammatory cytokine produced by mammalian cells, hi preferred embodiments, the proinflammatory cytokine is TNF, IL-lα, IL-1 β, MIF and/or IL-6, because those proinflammatory cytokines are particularly important mediators of disease. The methods of these embodiments are useful for in vitro applications, such as in studies for determining biological characteristics of proinflammatory cytokine production in cells. However, the preferred embodiments are in vivo therapeutic applications, where the cells are in a patient suffering from, or at risk for, a condition characterized by activation of an inflammatory cytokine cascade. h certain embodiments, the present invention is directed to a method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade. The method comprises administering to the patient any ofthe HMGB A box compositions (including non-naturally occurring A box polypeptides and A box biologically active fragments), any ofthe HMGB B box or B box biologically active fragment antibody compositions (including non-naturally occurring B box polypeptides or biologically active fragments thereof), and/or any of the combination therapy compositions discussed above. This method would be expected to be useful for any condition that is mediated by an inflammatory cytokine cascade, including any of those that have been previously enumerated. As with previously described in vivo methods, preferred conditions include appendicitis, peptic, gastric or duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute or ischemic colitis, hepatitis, Crohn's disease, asthma, allergy, anaphylactic shock, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, septic abortion, disseminated bacteremia, bums, Alzheimer's disease, cerebral infarction, cerebral embolism, spinal cord injury, paralysis, allograft rejection or graft- versus-host disease. In the most preferred embodiments, the condition is endotoxic shock or allograft rejection. Where the condition is allograft rejection, the composition may advantageously also include an immunosuppressant that is used to inhibit allograft rejection, such as cyclosporin.
These methods can also usefully include the administration of an antagonist of an early sepsis mediator, an anti-αvβ3 antibody, an anti JL-9 antibody, a B7 antagonist (e.g., CTLA4Ig, an anti-CD80 antibody, an anti-CD86 antibody), methotrexate, and/or a CD40 antagonist (e.g., anti-CD40 ligand (CD40L)). The nature of these agents has been previously discussed. The B box polypeptides and biologically active fragments thereof described herein can be used to induce inflammatory cytokines in the appropriate isolated cells in vitro, or ex vivo, or as a treatment in vivo. In any of these treatments, the polypeptide or fragment can be administered by providing a DNA or RNA vector encoding the B box or B box fragment, with the appropriate control sequences operably linked to the encoded B box or B box fragment, so that the B box or B box fragment is synthesized in the treated cell or patient. In vivo applications include the use ofthe B box polypeptides or B box fragment polypeptides or vectors as a weight loss treatment. See WO 00/47104 (the entire teachings of which are incorporated herein by reference), demonstrating that treatment with HMGBl induces weight loss. In certain embodiments, the present invention is directed to methods of stimulating the release of a proinflammatory cytokine from a cell. The method comprises treating the cell with any ofthe B box polypeptides or biologically active B box fragment polypeptides, for example, polypeptides that comprise or consist of the sequence of SEQ ID NO:5, SEQ ID NO:20, SEQ ID NO:58, SEQ ID NO: 16, or SEQ ID NO:23, as described herein (including non-naturally occurring B box polypeptides and fragments). This method is useful for in vitro applications, for example, for studying the effect of proinflammatory cytokine production on the biology ofthe producing cell. Since the HMGB B box has the activity ofthe HMGB protein, the B box would also be expected to induce weight loss. Therefore, in additional embodiments, the present invention is a method for effecting weight loss or treating obesity in a patient. The method comprises administering to the patient an effective amount of any ofthe B box polypeptides or B box fragment polypeptides described herein (including non-naturally occurring B box polypeptides and fragments). In another embodiment, the B box polypeptide or B box fragment polypeptide is in a pharmaceutically acceptable carrier.
Screening for Modulators ofthe Release of Proinflammatory Cytokines from Cells The present invention is also directed to a method of determining whether a compound (test compound) inhibits inflammation and/or an inflammatory response. The method comprises combining the compound with (a) a cell that releases a proinflammatory cytokine when exposed to a vertebrate HMGB B box or a biologically active fragment thereof, and (b) the HMGB B box or a biologically active fragment thereof, and then determining whether the compound inhibits the release ofthe proinflammatory cytokine from the cell, as compared to a suitable control. A compound that inhibits the release ofthe proinflammatory cytokine in this assay is a compound that can be used to treat inflammation and/or an inflammatory response. The HMGB B box or biologically active HMGB B box fragment can be endogenous to the cell or can be introduced into the cell using standard recombinant molecular biology techniques. Any cell that releases a proinflammatory cytokine in response to exposure to a vertebrate HMGB B box or biologically active fragment thereof in the absence of a test compound would be expected to be useful for this invention. It is envisioned that the cell that is selected would be important in the etiology ofthe condition to be treated with the inhibitory compound that is being tested. For many conditions, it is expected that the preferred cell is a human macrophage.
Any method for determining whether the compound inhibits the release ofthe proinflammatory cytokine from the cell would be useful for these embodiments. It is envisioned that the preferred methods are the direct measurement ofthe proinflammatory cytokine, for example, with any of a number of commercially available ELISA assays. However, in some embodiments, the measurement ofthe inflammatory effect of released cytokines may be preferable, particularly when there are several proinflammatory cytokines produced by the test cell. As previously discussed, for many important disorders, the predominant proinflammatory cytokines are TNF, IL-lα, IL-lβ, MIF or IL-6; particularly TNF. The present invention also features a method of determining whether a compound increases an inflammatory response and/or inflammation. The method comprises combining the compound (test compound) with (a) a cell that releases a proinflammatory cytokine when exposed to a vertebrate HMGB A box or a biologically active fragment thereof, and (b) the HMGB A box or biologically active fragment, and then determining whether the compound increases the release ofthe proinflammatory cytoldne from the cell, as compared to a suitable control. A compound that increases the release ofthe proinflammatory cytokine in this assay is a compound that can be used to increase an inflammatory response and/or inflammation. The HMGB A box or HMGB A box biologically active fragment can be endogenous to the cell or can be introduced into the cell using standard recombinant molecular biology techniques.
Similar to the cell types useful for identifying inhibitors of inflammation described above, any cell in which release of a proinflammatory cytokine is normally inhibited in response to exposure to a vertebrate HMGB A box or a biologically active fragment thereof in the absence of any test compound would be expected to be useful for this invention. It is envisioned that the cell that is selected would be important in the etiology ofthe condition to be treated with the inhibitory compound that is being tested. For many conditions, it is expected that the preferred cell is a human macrophage.
Any method for determining whether the compound increases the release of the proinflammatory cytokine from the cell would be useful for these embodiments. It is envisioned that the preferred methods are the direct measurement ofthe proinflammatory cytokine, for example, with any of a number of commercially available ELISA assays. However, in some embodiments, the measurement ofthe inflammatory effect of released cytokines may be preferable, particularly when there are several proinflammatory cytokines produced by the test cell. As previously discussed, for many important disorders, the predominant proinflammatory cytokines are TNF, IL-lα, IL-lβ, MIF or IL-6; particularly TNF.
Preferred embodiments ofthe invention are described in the following examples. Other embodiments within the scope ofthe invention will be apparent to one skilled in the art from consideration ofthe specification or practice ofthe invention as disclosed herein. It is intended that the specification, together with the examples and claims, be considered exemplary only. Example 1 : Materials and Methods
Cloning of HMGBl and Production of HMGBl Mutants The following methods were used to prepare clones and mutants of human HMGBl . Recombinant full length human HMGBl (651 base pairs; GenBank Accession Number U51677) was cloned by PCR amplification from a human brain Quick-Clone cDNA preparation (Clontech, Palo Alto, CA) using the following primers; forward primer: 5' GATGGGCAAAGGAGATCCTAAG 3' (SEQ ID NO:6) and reverse primer: 5* GCGGCCGCTTATTCATCATCATCATCTTC 3' (SEQ ID NO:7). Human HMGBl mutants were cloned and purified as follows. A truncated form of human HMGB 1 was cloned by PCR amplification from a Human Brain Quick-Clone cDNA preparation (Clontech, Palo Alto, CA). The primers used were (forward and reverse, respectively):
Carboxy terminus mutant (557 bp): 5' GATGGGCAAAGGAGATCCTAAG 3' (SEQ ID NO:8) and 5' GCGGCCGC TCACTTGCTTTTTTCAGCCTTGAC 3' (SEQ ID NO:9).
Amino terminus+B box mutant (486 bp): 5' GAGCATAAGAAGAAGCACCCA 3' (SEQ ID NO: 10) and 5* GCGGCCGC TCACTTGCTTTTTTCAGCCTTGAC 3' (SEQ ID NO: 11).
B box mutant (233 bp): 5' AAGTTCAAGGATCCCAATGCAAAG 3' (SEQ ID NO: 12) and 5' GCGGCCGCTCAATATGCAGCTATATCCTTTTC 3' (SEQ ID NO:13).
Amino terminus+A box mutant (261 bp): 5' GATGGGCAAAGGAGATCCTAAG 3' (SEQ ID NO: 14) and 5' TCACTTTTTTGTCTCCCCTTTGGG 3' (SEQ ID NO: 15).
A stop codon was added to each mutant to ensure the accuracy of protein size. PCR products were subcloned into pCRII-TOPO vector EcoRI sites using the TA cloning method per manufacturer's instruction (Invitrogen, Carlsbad, CA). After amplification, the PCR product was digested with EcoRI and subcloned into an expression vector with a GST tag pGEX (Pharmacia); correct orientation and positive clones were confirmed by DNA sequencing on both strands. The recombinant plasmids were transformed into protease deficient E. coli strains BL21 or BL21 (DE3)plysS (Novagen, Madison, WI) and fusion protein expression was induced by isopropyl-D-thiogalactopyranoside (IPTG). Recombinant proteins were obtained using affinity purification with the glutathione Sepharose resin column (Pharmacia).
The HMGB mutants generated as described above have the following amino acid sequences:
Wild type HMGBl:
MGKGDPKKPTGKMSSYAFFVQTCREEHKKKHPDASVNFSEFSKKCSERWKT MSAKEKGl^EDMAKADKARYEREMKTYIPPKGETKKKFKDPNAPKRLPSAF FLFCSEYRPKIKGEHPGLSIGDVAKKLGEMWNNTAADDKQPYEKKAAKLKE KYEKDIAAYRAKGKPD AAKKGVVKAEKSKKKKEEEEDEEDEEDEEEEEDEE DEEDEEEDDDDE (SEQ ID NO: 18)
Carboxy terminus mutant:
MGKGDPKKPTGKMSSYAFFVQTCREEHKKKHPDASVNFSEFSKKCSERWKT MSAKΕKGKFEDMAKADKARYEREMKTYIPPKGETKKKFKDPNAPKRLPSAF FLFCSEYRPKIKGEHPGLSIGDVAKKLGEMWNNTAADDKQPYEKKAAKLKE KYEKDIAAYRAKGKPD AAKKGVVKAEKSK (SEQ ID NO: 19)
B Box mutant: FKDPNAPKRLPSAFFLFCSEYRPKIKGEHPGLSIGDNAKKLGEM WΝΝTAADDKQPYEKKAAKLKEKYEKDIAAY (SEQ ID NO: 20)
Amino terminus + A Box mutant: MGKGDPKKPTGKMSSYAFFNQTCREEHKKK HPDASVΝFSEFSKKCSERWKTMSAKEKGKFEDMAKADKARYEREMKTYIPP KGET (SEQ ID NO: 21), wherein the A box consists ofthe sequence PTGKMSSYAFFNQTCREEHKKKHPDASNNFSEFSKKCSERWKTMSAKEKGK FEDMAKADKARYEREMKTYIPPKGET (SEQ ID NO:22)
A polypeptide generated from a GST vector lacking HMGBl protein was included as a control (containing a GST tag only). To inactive the bacterial DNA that bound to the wild type HMGBl and some ofthe mutants (carboxy terminus and B box), DNase I (Life Technologies), for carboxy terminus and B box mutants, or benzonase nuclease (Novagen, Madison, WI), for wild type HMGBl, was added at about 20 units/ml bacteria lysate. Degradation of DNA was verified by ethidium bromide staining ofthe agarose gel containing HMGBl proteins before and after the treatment. The protein eluates were passed over a polymyxin B column (Pierce, Rockford, IL) to remove any contaminating LPS, and dialyzed extensively against phosphate buffered saline to remove excess reduced glutathione. The preparations were then lyophilized and redissolved in sterile water before use. LPS levels were less than 60 pg/μg protein for all ofthe mutants and 300 pg/μg for wild type HMG-1, as measured by Limulus amebocyte lysate assay (Bio Whittaker Inc., Walkersville, MD). The integrity of protein was verified by SDS-PAGE. Recombinant rat HMGBl (Wang et al., Science 285: 248-251, 1999) was used in some experiments since it does not have degraded fragments as observed in purified human HMGBl .
Peptide Synthesis Peptides were synthesized and HPLC purified at Utah State University
Biotechnology Center (Logan, Utah) at 90% purity. Endotoxin was not detectable in the synthetic peptide preparations as measured by Limulus assay.
Cell Culture
Murine macrophage-like RAW 264.7 cells (American Type Culture Collection, Rockville, MD) were cultured in RPMI 1640 medium (Life
Technologies, Grand Island NY) supplemented with 10%> fetal bovine serum (Gemini, Catabasas, CA), penicillin and streptomycin (Life Technologies) and were used at 90%) confluence in serum-free Opti-MEM I medium (Life Technologies, Grand Island, NY). Polymyxin B (Sigma, St. Louis, MO) was routinely added at 100-1,000 units/ml to neutralize the activity of any contaminating LPS as previously described; polymyxin B alone did not influence cell viability assessed with trypan blue (Wang et al., supra). Polymyxin B was not used in experiments of synthetic peptide studies.
Measurement of TNF Release From Cells
TNF release was measured by a standard murine fibroblast L929 (ATCC, American Type Culture Collection, Rockville, MD) cytotoxicity bioassay (Bianchi et al., Journal of Experimental Medicine 183:927-936, 1996) with the minimum detectable concentration of 30 pg/ml. Recombinant mouse TNF was obtained from R&D system Inc., (Minneapolis, MN). Murine fibroblast L929 cells (ATCC) were cultured in DMEM (Life Technologies, Grand Island, NY) supplemented with 10% fetal bovine serum (Gemini, Catabasas, CA), penicillin (50 units/ml) and streptomycin (50 μg/ml) (Life Technologies) in a humidified incubator with 5% CO2.
Antibody Production
Polyclonal antibodies against HMGBl B box were raised in rabbits (Cocalico Biologicals, Inc., Reamstown, PA) and assayed for titer by immunoblotting. IgG was purified from anti-HMGB 1 antiserum using Protein A agarose according to manufacturer's instructions (Pierce, Rockford, IL). Anti-HMGB 1 B box antibodies were affinity purified using cyanogen bromide activated Sepharose beads (Cocalico Biological, Inc.). Non-immune rabbit IgG was purchased from Sigma (St. Louis, MO). Antibodies detected full length HMGBl and B box in immunoassay, but did not cross react with TNF, IL-1 and IL-6.
Labeling of HMGBl with Na-'25I and cell surface binding Purified HMGBl protein (10 μg) was radiolabeled with 0.2 mCi of carrier- free 125I (NEN Life Science Products Inc., Boston, MA) using Iodo-beads (Pierce, Rockford, IL) according to the manufacturer's instructions. 125I-HMGB1 protein was separated from un-reacted 125I by gel chromatography columns (P6 Micro Bio-Spin Chromatography Columns, Bio-Rad Laboratories, Hercules, CA) previously equilibrated with 300 mM sodium chloride, 17.5 mM sodium citrate, pH 7.0, and 0.1%) bovine serum albumin (BSA). The specific activity ofthe eluted HMGBl was about 2.8 x 106 cpm/μg protein. Cell surface binding studies were performed as previously described (Yang et al., Am. J. Physiol. 275:C675-C683, 1998). RAW 264.7 cells were plated on 24-well dishes and grown to confluence. Cells were washed twice with ice-cold PBS containing 0.1 % BSA and binding was carried out at 4°C for 2 hours with 0.5 ml binding buffer containing 120 mM sodium chloride, 1.2 mM magnesium sulfate, 15 mM sodium acetate, 5 mM potassium chloride, 10 mM Tris.HCl, pH 7.4, 0.2% BSA, 5mM glucose and 25,000 cpm 125I-HMGB1. At the end ofthe incubation the supernatants were discarded and the cells were washed three times with 0.5 ml of ice-cold PBS with 0.1% BSA and lysed with 0.5 ml of 0.5 N NaOH and 0.1% SDS for 20 minutes at room temperature. The radioactivity in the lysate was then measured using a gamma counter. Specific binding was determined as total binding minus the radioactivity obtained in the presence of an excess amount of unlabeled HMGB 1 or A box proteins.
Animal Experiments
TNF knock out mice were obtained from Amgen (Thousand Oaks, CA) and were on a B6xl29 background. Age-matched wild-type B6xl29 mice were used as a control for the studies. Mice were bred in-house at the University of Florida specific pathogen-free transgenic mouse facility (Gainesville, FL) and were used at 6-8 weeks of age.
Male 6-8 week old Balb/c and C3H/HeJ mice were purchased from Harlen Sprague-Dawley (Indianapolis, IN) and were allowed to acclimate for 7 days before use in experiments. All animals were housed in the North Shore University Hospital Animal Facility under standard temperature, and a light and dark cycle.
Cecal Ligation and Puncture
Cecal ligation and puncture (CLP) was performed as described previously (Fink and Heard, J. Surg. Res. 49:186-196, 1990; Wichmann et al, Crit. Care Med. 26:2078-2086, 1998; and Remick et al, Shock 4:89-95, 1995). Briefly, Balb/c mice were anesthetized with 75 mg/kg ketamine (Fort Dodge, Fort Dodge, Iowa) and 20 mg/kg of xylazine (Bohringer Ingelheim, St. Joseph, MO) intramuscularly. A midline incision was performed, and the cecum was isolated. A 6-0 prolene suture ligature was placed at a level 5.0 mm from the cecal tip away from the ileocecal valve.
The ligated cecal stump was then punctured once with a 22-gauge needle, without direct extrusion of stool. The cecum was then placed back into its normal intra-abdominal position. The abdomen was then closed with a running suture of 6-0 prolene in two layers, peritoneum and fascia separately to prevent leakage of fluid. All animals were resuscitated with a normal saline solution administered sub- cutaneously at 20 ml/kg of body weight. Each mouse received a subcutaneous injection of imipenem (0.5 mg/mouse) (Primaxin, Merck & Co., Inc., West Point, PA) 30 minutes after the surgery. Animals were then allowed to recuperate. Mortality was recorded for up to 1 week after the procedure; survivors were followed for 2 weeks to ensure no late mortalities had occurred.
D-galactosamine Sensitized Mice
The D-galactosamine-sensitized model has been described previously (Galanos et al., Proc Natl. Acad. Sci. USA 76: 5939-5943, 1979; and Lehmann et al., J. Exp. Med. 165: 657-663, 1997). Mice were injected intraperitoneally with 20 mg D-galactosamine-HCL (Sigma)/mouse (in 200. μl PBS) and 0.1 or 1 mg of either HMBG1 B box or vector protein (in 200 μl PBS). Mortality was recorded daily for up to 72 hours after injection; survivors were followed for 2 weeks, and no later deaths from B box toxicity were observed.
Spleen bacteria culture
Fourteen mice received either anti-HMGB 1 antibody (n=7) or control (n=7) at 24 and 30 hours after CLP, as described herein, and were euthanized for necropsy. Spleen bacteria were recovered as described previously (Villa et al., J. Endotoxin Res. 4:197-204, 1997). Spleens were removed using sterile technique and homogenized in 2 ml of PBS. After serial dilutions with PBS, the homogenate was plated as 0.15 ml aliquots on tryptic soy agar plates (Difco, Detroit, MI) and CFU were counted after overnight incubation at 37°C.
Statistical Analysis Data are presented as mean ± SEM unless otherwise stated. Differences between groups were determined by two-tailed Student's t-test, one-way ANOVA followed by the least significant difference test or 2 tailed Fisher's Exact Test.
Example 2: Mapping the HMGBl Domains for Promotion of Cytokine Activity HMGBl has 2 folded DNA binding domains (A and B boxes) and a negatively-charged acidic carboxyl tail. To elucidate the structural basis of HMGBl cytokine activity, and to map the inflammatory protein domain, we expressed full length and truncated forms of HMGBl by mutagenesis and screened the purified proteins for stimulating activity in monocyte cultures (FIG. 1). Full length HMGBl, a mutant in which the carboxy terminus was deleted, a mutant containing only the B box, and a mutant containing only the A box were generated. These mutants of human HMGBl were made by polymerase chain reaction (PCR) using specific primers as described herein, and the mutant proteins were expressed using a glutathione S-transferase (GST) gene fusion system (Pharmacia Biotech, Piscataway, NJ) in accordance with the manufacturer's instructions. Briefly, DNA fragments, made by PCR methods, were fused to GST fusion vectors and amplified in E. coli. The expressed HMGBl protein and HMGBl mutants were then isolated using a GST affinity column.
The effect ofthe mutants on TNF release from Murine macrophage-like RAW 264.7 cells (ATCC) was carried out as follows. RAW 264.7 cells were cultured in RPMI 1640 medium (Life Technologies, Grand Island NY) supplemented with 10% fetal bovine serum (Gemini, Catabasas, CA), penicillin and streptomycin (Life Technologies). Polymyxin (Sigma, St. Louis, MO) was added at 100 units/ml to suppress the activity of any contaminating LPS. Cells were incubated with 1 μg/ml of full length (wild-type) HMGBl and each HMGBl mutant protein in Opti- MEM I medium for 8 hours. Conditioned supernatants (containing TNF which had been released from the cells) were collected and TNF released from the cells was measured by a standard murine fibroblast L929 (ATCC) cytotoxicity bioassay (Bianchi et al., supra) with the minimum detectable concentration of 30 pg/ml. Recombinant mouse TNF was obtained from R & D Systems Inc., (Minneapolis, MN) and used as control in these experiments. The results of this study are shown in FIG. 1. Data in FIG. 1 are all presented as mean + SEM unless otherwise indicated. (N=6-10).
As shown in FIG. 1, wild-type HMGBl and carboxy 1-truncated HMGBl significantly stimulated TNF release by monocyte cultures (murine macrophage-like RAW 264.7 cells). The B box was a potent activator of monocyte TNF release. This stimulating effect ofthe B box was specific, because A box only weakly activated TNF release.
Example 3: HMGBl B Box Protein Promotes Cytokine Activity in a Dose Dependent Manner
To further examine the effect of HMGBl B box on cytokine production, varying amounts of HMGBl B box were evaluated for the effects on TNF, IL-1B, and IL-6 production in murine macrophage-like RAW 264.7 cells. RAW 264.7 cells were stimulated with B box protein at 0-10 μg/ml, as indicated in FIGS. 2A-2C for 8 hours. Conditioned media were harvested and measured for TNF, IL-lβ and IL-6 levels. TNF levels were measured as described herein, and IL-lβ and IL-6 levels were measured using the mouse IL-lβ and IL-6 enzyme-linked immunosorbent assay (ELISA) kits (R&D System Inc., Minneapolis, MN) and N>5 for all experiments. The results ofthe studies are shown in FIGS. 2A-2C. As shown in FIG. 2A, TNF release from RAW 264.7 cells increased with increased amounts of B box administered to the cells. As shown in FIG. 2B, addition of 1 μg/ml or 10 μg/ml of B box resulted in increased release of IL-lβ from RAW 264.7 cells. In addition, as shown in FIG. 2C, IL-6 release from RAW 264.7 cells increased with increased amounts of B box administered to the cells. The kinetics of B box-induced TNF release were also examined. TNF release and TNF mRNA expression were measured in RAW 264.7 cells induced by B box polypeptide or GST tag polypeptide only used as a control (vector) (10 μg/ml) for 0 to 48 hours. Supernatants were analyzed for TNF protein levels by an L929 cytotoxicity assay (N=3-5) as described herein. For mRNA measurement, cells were plated in 100 mm plates and treated in Opti-MEM I medium containing B box polypeptide or the vector alone for 0, 4, 8, or 24 hours, as indicated in FIG. 2D. The vector only sample was assayed at the 4 hour time point. Cells were scraped off the plate and total RNA was isolated using the RNAzol B method in accordance with the manufacturer's instructions (Tel-Test "B", Inc., Friendswood, TX). TNF (287 bp) was measured by RNase protection assay (Ambion, Austin, TX). Equal loading and the integrity of RNA was verified by ethidium bromide staining ofthe RNA sample on an agarose-formaldehyde gel. The results ofthe RNase protection assay are shown in FIG. 2D. As shown in FIG. 2D, B box activation of monocytes occurred at the level of gene transcription, because TNF mRNA was increased significantly in monocytes exposed to B box protein (FIG. 2B). TNF mRNA expression was maximal at 4 hours and decreased at 8 and 24 hours. The vector only control (GST tag) showed no effect on TNF mRNA expression. A similar study was carried out measuring TNF protein released from RAW 264.7 cells 0, 4, 8, 24, 32 or 48 hours after administration of B box or vector only (GST tag), using the L929 cytotoxicity assay described herein. Compared to the control (medium only), B box treatment stimulated TNF protein expression (FIG. 2E) and vector alone (FIG. 2F) did not. Data are representative of three separate experiments. Together these data indicate that the HMGB 1 B box domain has cytokine activity and is responsible for the cytokine stimulating activity of full length HMGB 1.
In summary, the HMGBl B box dose-dependently stimulated release of TNF, IL-lβ and IL-6 from monocyte cultures (FIGS. 2A-2C), in agreement with the inflammatory activity of full length HMGBl (Andersson et al., J. Exp. Med. 192: 565-570, 2000). In addition, these studies indicate that maximum TNF protein release occurred within 8 hours (FIG. 2E). This delayed pattern of TNF release is similar to TNF release induced by HMGBl itself, and is significantly later than the kinetics of TNF induced by LPS (Andersson et al, supra).
Example 4: The First 20 Amino Acids ofthe HMGBl B Box Stimulate TNF Activity The TNF-stimulating activity ofthe HMGBl B box was further mapped. This study was carried out as follows. Fragments ofthe B box were generated using synthetic peptide protection techniques, as described herein. Five HMGBl B box fragments (from SEQ ID NO:20), containing amino acids 1-20, 16-25, 30-49, 45-64, or 60-74 ofthe HMGBl B box were generated, as indicated in FIG. 3. RAW 264.7 cells were treated with B box (1 μg/ml) or a synthetic peptide fragment ofthe B box (10 μg/ml), as indicated in FIG. 3, for 10 hours and TNF release in the supematants was measured as described herein. Data shown are mean ± SEM, (n=3 experiments, each done in duplicate and validated using 3 separate lots of synthetic peptides). As shown in FIG. 3, TNF-stimulating activity was retained by a synthetic peptide corresponding to amino acids 1-20 ofthe HMGBl B box of SEQ ID NO:20 (flcdpnaplcrlpsafflfcse; SEQ ID NO :23). The TNF stimulating activity of the 1 -20- mer was less potent than either the full length synthetic B box (1-74-mer), or full length HMGBl, but the stimulatory effects were specific because the synthetic 20- mers for amino acid fragments containing 16-25, 30-49, 45-64, or 60-74 ofthe HMGBl B box did not induce TNF release. These results are direct evidence that the macrophage stimulating activity ofthe B box specifically maps to the first 20 amino acids ofthe HMGB B box domain of SEQ ID NO:20). This B box fragment can be used in the same manner as a polypeptide encoding a full length B box polypeptide, for example, to stimulate release of a proinflammatory cytokine, or to treat a condition in a patient characterized by activation of an inflammatory cytokine cascade.
Example 5: HMGBl A Box Protein Antagonizes HMGBl Induced Cytokine Activity in a Dose Dependent Manner
Weak agonists are by definition antagonists. Since the HMGBl A box only weakly induced TNF production, as shown in FIG. 1, the ability of HMGBl A box to act as an antagonist of HMGBl activity was evaluated. This study was carried out as follows. Sub-confluent RAW 264.7 cells in 24-well dishes were treated with HMGBl (1 μg/ml) and 0, 5, 10, or 25 μg/ml of A box for 16 hours in Opti-MEM I medium in the presence of polymyxin B (100 units/ml). The TNF-stimulating activity (assayed using the L929 cytotoxicity assay described herein) in the sample receiving no A box was expressed as 100%, and the inhibition by A box was expressed as percent of HMGBl alone. The results ofthe effect of A box on TNF release from RAW 264.7 cells is shown in FIG. 4A. As shown in FIG. 4A, the A box dose-dependently inhibited HMGBl induced TNF release with an apparent EC50 of approximately 7.5 μg/ml. Data in FIG. 4A are presented as mean ± SD (n= 2-3 independent experiments).
Example 6: HMGBl A Box Protein Inhibits Full Length HMGBl and HMGBl B Box Cytokine Activity
Antagonism of full length HMGBl activity by HMGBl A box or GST tag (vector control) was also determined by measuring TNF release from RAW 264.7 macrophage cultures stimulated by co-addition of A box with full length HMGBl. RAW 264.7 macrophage cells (ATCC) were seeded into 24-well tissue culture plates and used at 90% confluence. The cells were treated with HMGBl, and/or A boxes as indicated for 16 hours in Optimum I medium (Life Technologies, Grand Island, NY) in the presence of polymyxin B (100 units/ml, Sigma, St. Louis, MO) and supematants were collected for TNF measurement (mouse ELISA kit from R&D System Inc, Minneapolis, MN). TNF-inducing activity was expressed as a percentage ofthe activity achieved with HMGBl alone. The results of these studies are shown in FIG. 4B. FIG. 4B is a histogram ofthe effect of HMGBl (HMG-1), alone, A box alone, Vector (control) alone, HMGBl in combination with A box, and HMGBl in combination with vector. As shown in FIG. 4B, HMGBl A box significantly attenuated the TNF stimulating activity of full length HMGBl. Example 7: HMGBl A Box Protein Inhibits HMGBl Cytokine Activity by Binding to It
To determine whether the HMGBl A box acts as an antagonist by displacing HMGBl binding, 125I-labeled-HMGBl was added to macrophage cultures and binding was measured at 4°C after 2 hours. Binding assays in RAW 264.7 cells were performed as described herein. 125I-HMGB1 binding was measured in RAW 264.7 cells plated in 24-well dishes for the times indicated in FIG. 5A. Specific binding shown equals total cell-associated 125I-HMGB1 (CPM/well) minus cell associated CPM/well in the presence of 5,000 fold molar excess of unlabeled HMGBl. FIG. 5 A is a graph ofthe binding of 125I-HMGB1 over time. As shown in FIG. 5 A,
HMGBl exhibited saturable first order binding kinetics. The specificity of binding was assessed as described in Example 1.
In addition, 125I-HMG-1 binding was measured in RAW 264.7 cells plated on 24-well dishes and incubated with 125I HMGBl alone or in the presence of unlabeled HMGBl or A box. The results of this binding assay are shown in FIG. 5B. Data represents mean ± SEM from 3 separate experiments. FIG. 5B is a histogram ofthe cell surface binding of 125I-HMGB1 in the absence of unlabeled HMGBl or HMGBl A box, or in the presence of 5,000 molar excess of unlabeled HMGBl or HMGBl A box, measured as a percent ofthe total CPM/well. In FIG. 5B, "Total" equals counts per minutes (CPMVwell of cell associated 125I-HMGB1 in the absence of unlabeled HMGBl or A box for 2 hours at 4°C. "HMGBl" or "A box" equals CPM/well of cell-associated 125I-HMGB1 in the presence of 5,000 molar excess of unlabeled HMGBl or unlabeled A box. The data are expressed as the percent of total counts obtained in the absence of unlabeled HMGBl proteins (2,382,179 CPM/well). These results indicate that the HMGB 1 A box is a competitive antagonist of HMGB 1 activity in vitro and inhibits the TNF-stimulating activity of HMGBl.
Example 8: Inhibition of Full Length HMGBl and HMGBl B Box Cytokine Activity by Anti-B Box Polyclonal Antibodies.
The ability of antibodies directed against the HMGBl B box to modulated the effect of full length or HMGB 1 B box was also assessed. Affinity purified antibodies directed against the HMGBl B box (B box antibodies) were generated as described herein and using standard techniques. To assay the effect ofthe antibodies on HMGBl -induced or HMGBl B box-induced TNF release from RAW 264.7 cells, sub-confluent RAW 264.7 cells in 24-well dishes were treated with HMG-1 (1 μg/ml) or HMGB 1 B box (10 μg/ml) for 10 hours with or without anti-B box antibody (25 μg/ml or 100 μg/ml antigen affinity purified, Cocalico Biologicals, Inc., Reamstown, PA) or non-immune IgG (25 μg/ml or 100 μg/ml; Sigma) added. TNF release from the RAW 264.7 cells was measured using the L929 cytotoxicity assay as described herein. The results of this study are shown in FIG. 6, which is a histogram of TNF released by RAW 264.7 cells administered nothing, 1 μg/ml of HMGB 1 , 1 μg/ml of HMGBl plus 25 μg/ml of anti-B box antibody, 1 μg/ml of HMGBl plus 25 μg/ml of IgG (control), 10 μg/ml of B-box, 10 μg/ml of B-box plus 100 μg/ml of anti-B box antibody or 10 μg/ml of B-box plus 100 μg/ml of IgG (control). The amount of TNF released from the cells induced by HMGBl alone (without addition of B box antibodies) was set as 100%, and the data shown in FIG. 6 are the results of 3 independent experiments. As shown in FIG. 6, affinity purified antibodies directed against the HMGBl B box significantly inhibited TNF release induced by either full length HMGBl or the HMGBl B box. These results indicate that such an antibody can be used to modulate HMGBl function.
Example 9: HMGBl B Box Protein is Toxic to D-galactosamine-sensitized Balb/c Mice
To investigate whether the HMGBl B box has cytokine activity in vivo, we administered HMGBl B box protein to unanesthetized Balb/c mice sensitized with D-galactosamine (D-gal), a model that is widely used to study cytokine toxicity (Galanos et al, supra). Briefly, mice (20-25 grams, male, Harlan Sprague-Dawley, Indianapolis, IN) were intraperitoneally injected with D-gal (20 mg) (Sigma, St. Louis, Missouri) and B box (0.1 mg/ml/mouse or 1 mg/ml/mouse) or GST tag (vector; 0.1 mg/ml/mouse or 1 mg/ml/mouse), as indicated in Table 1. Survival of the mice was monitored up to 7 days to ensure no late death occurred. The results of this study are shown in Table 1. Table 1: Toxicity of HMGBl B box on D-galactosamine-sensitized Balb/c Mice
Figure imgf000060_0001
*P<0.01 versus vector alone as tested by Fisher's Exact Test
The results of this study showed that the HMGBl B box was lethal to D- galactosamine-sensitized mice in a dose-dependent mamier. In all instances in which death occurred, it occurred within 12 hours. Lethality was not observed in mice treated with comparable preparations ofthe purified GST vector protein devoid of B box.
Example 10: Histology of D-galactosamine-sensitized Balb/c Mice or C3H/HeJ Mice Administered HMGBl B Box Protein
To further assess the lethality ofthe HMGBl B box protein in vivo the HMGBl B box was again administered to D-galactosamine-sensitized Balb/c mice. Mice (3 per group) received D-gal (20 mg/mouse) plus B box or vector (1 mg/mouse) intraperitoneally for 7 hours and were then sacrificed by decapitation. Blood was collected, and organs (liver, heart, kidney and lung) were harvested and fixed in 10% formaldehyde. Tissue sections were prepared with hematoxylin and eosin staining for histological evaluation (Criterion Inc., Vancouver, Canada). The results of these studies are shown in FIGS. 7A-7J, which are scanned images of hematoxylin and eosin stained kidney sections (FIG. 7A), myocardium sections (FIG. 7C), lung sections (FIG. 7E), and liver sections (FIGS. 7G and 71) obtained from an untreated mouse and kidney sections (FIG. 7B), myocardium sections (FIG. 7D), lung sections (FIG. 7F), and liver sections (FIGS. 7H and 7J) obtained from mice treated with the HMGB 1 B box. Compared to the control mice, B box treatment caused no abnormality in kidneys (FIGS. 7A and 7B) and lungs (FIGS. 7E and 7F). The mice had some ischemic changes and loss of cross striation in myocardial fibers in the heart (FIGS. 7C and 7D as indicated by the arrow in FIG. 7D). Liver showed most ofthe damage by the B box as illustrated by active hepatitis (FIGS. 7G-7J). In FIG. 7J, hepatocyte dropouts are seen surrounded by accumulated polymorphonuclear leukocytes. The arrows in FIG. 7J point to the sites of polymorphonuclear accumulation (dotted) or apoptotic hepatocytes (solid). Administration of HMGBl B box in vivo also stimulated significantly increased serum levels of IL-6 (315+93 vs.20+7 pg/ml, B box vs. control, p<0.05) and IL-lβ (15+3 vs. 4+1 pg/ml, B box vs. control, p<0.05).
Administration of B box protein to C3H/HeJ mice (which do not respond to endotoxin) was also lethal, indicating that HMGBl B box is lethal in the absence of LPS signal transduction. Hematoxylin and eosin stained sections of lung and kidney collected 8 hours after administration of B box revealed no abnormal morphologic changes. Examination of sections from the heart however, revealed evidence of ischemia with loss of cross striation associated with amorphous pink cytoplasm in myocardial fibers. Sections from liver showed mild acute inflammatory responses, with some hepatocyte dropout and apoptosis, and occasional polymorphonuclear leukocytes. These specific pathological changes were comparable to those observed after administration of full length HMGB 1 and confirm that the B box alone can recapitulate the lethal pathological response to HMGBl in vivo.
To address whether the TNF-stimulating activity of HMGBl contributes to the mediation of lethality by B box, we measured lethality in TNF knock-out mice (TNF-KO, Nowak et al., Am. J. Physiol. Regul. Integr. Comp. Physiol. 278: R1202- R1209, 2000) and the wild-type controls (B6xl29 strain) sensitized with D- galactosamine (20 mg/mouse) and exposed to B box (1 mg/mouse, injected intraperitoneally). The B box was highly lethal to the wild-type mice (6 dead out of nine exposed) but lethality was not observed in the TNF-KO mice treated with B box (0 dead out of 9 exposed, p<0.05 v. wild type). Together with the data from the RAW 264.7 macrophage cultures, described herein, these data now indicate that the B box of HMGBl confers specific TNF-stimulating cytokine activity. Example 11 : HMGBl Protein Level is Increased in Septic Mice
To examine the role of HMGBl in sepsis, we established sepsis in mice and measured serum HMGB 1 using a quantitative immunoassay described previously (Wang et al, supra). Mice were subjected to cecal ligation and puncture (CLP), a well characterized model of sepsis caused by perforating a surgically-created cecal diverticulum, that leads to polymicrobial peritonitis and sepsis (Fink and Heard, supra; Wichmann et al., supra; and Remick et al, supra). Serum levels of HMGBl were then measured (Wang et al., supra). FIG. 8 shows the results of this study in a graph that illustrates the levels of HMGBl in mice 0 hours, 8 hours, 18 hours, 24 hours, 48 hours, and 72 hours after subjection to CLP. As shown in FIG. 8, serum HMGBl levels were not significantly increased for the first eight hours after cecal perforation, then increased significantly after 18 hours (FIG. 8). Increased serum HMGBl remained at elevated plateau levels for at least 72 hours after CLP, a kinetic profile that is quite similar to the previously-described, delayed HMGB 1 kinetics in endotoxemia (Wang et al., supra). This temporal pattern of HMGBl release corresponded closely to the development of signs of sepsis in the mice. During the first eight hours after cecal perforation the animals were observed to be mildly ill, with some diminished activity and loss of exploratory behavior. Over the ensuing 18 hours the animals became gravely ill, huddled together in groups with piloerection, did not seek water or food, and became minimally responsive to external stimuli or being examined by the handler.
Example 12: Treatment of Septic Mice with HMGBl A Box Protein Increases Survival of Mice
To determine whether the HMGBl A box can inhibit the lethality of HMGBl during sepsis, mice were subjected to cecal perforation and treated by administration of A box beginning 24 hours after the onset of sepsis. CLP was performed on male Balb/c mice as described herein. Animals were randomly grouped, with 15-25 mice per group. The HMGBl A box (60 or 600 μg/mouse each time) or vector (GST tag, 600 μg/mouse) alone was administered intraperitoneally twice daily for 3 days beginning 24 hours after CLP. Survival was monitored twice daily for up to 2 weeks to ensure no late death occurred. The results of this study are illustrated in FIG. 9, which is a graph ofthe effect of vector (GST; control) 60 μg/mouse or 600 μg/mouse on survival over time (*P<0.03 vs. control as tested by Fisher's exact test). As shown in FIG. 9, administration ofthe HMGBl A box significantly rescued mice from the lethal effects of sepsis, and improved survival from 28% in the animals treated with protein purified from the vector protein (GST) devoid of the A box, to 68%) in animals receiving A box (P<0.03 by Fischer's exact test). The rescuing effects ofthe HMGBl A box in this sepsis model were A box dose-dependent; animals treated with 600 μg/mouse of A box were observed to be significantly more alert, active, and to resume feeding behavior as compared to either control animals treated with vector-derived preparations, or to animals treated with only 60 μg A box. The latter animals remained gravely ill, with depressed activity and feeding for several days, and most died.
Example 13 : Treatment of Septic Mice with Anti-HMGB 1 Antibody Increases Survival of Mice
Passive immunization of critically ill septic mice with anti-HMGB 1 antibodies was also assessed. In this study, male Balb/c mice (20-25 gm) were subjected to CLP, as described herein. Affinity purified anti-HMGB 1 B box polyclonal antibody or rabbit IgG (as control) was administered at 600 μg/mouse beginning 24 hours after the surgery, and twice daily for 3 days. Survival was monitored for 2 weeks. The results of this study are shown in FIG. 10 A, which is a graph ofthe survival of septic mice treated with either a control antibody or an anti- HMGB 1 antibody. The results show that anti-HMGB 1 antibodies administered to the mice 24 hours after the onset of cecal perforation significantly rescued animals from death as compared to administration of non-immune antibodies (p<0.02 by Fisher's exact test). Within 12 hours after administration of anti-HMGB 1 antibodies, treated animals showed increased activity and responsiveness as compared to controls receiving non-immune antibodies. Whereas animals treated with non- immune antibodies remained huddled, ill kempt, and inactive, the treated animals improved significantly and within 48 hours resumed normal feeding behavior. Anti- HMGB 1 antibodies did not suppress bacterial proliferation in this model, because we observed comparable bacterial counts (CFU, the aerobic colony forming units) from spleen 31 hours after CLP in the treated animals as compared to animals receiving irrelevant antibodies (control bacteria counts = 3.5+0.9x104 CFU/g; n=7). Animals were monitored for up to 2 weeks afterwards, and late deaths were not observed, indicating that treatment with anti-HMGB 1 conferred complete rescue from lethal sepsis, and did not merely delay death.
To our knowledge, no other specific cytokine-directed therapeutic is as effective when administered so late after the onset of sepsis. By comparison, administration of anti-TNF actually increases mortality in this model, and anti-MIF antibodies are ineffective if administered more than 8 hours after cecal perforation (Remick et al, supra; and Calandra et al., Nature Med. 6:164-170, 2000). These data demonstrate that HMGBl can be targeted as late as 24 hours after cecal perforation in order to rescue lethal cases of established sepsis.
In another example ofthe rescue of endotoxemic mice using anti-B box antibodies, anti-HMGB 1 B box antibodies were evaluated for their ability to rescue LPS-induced septic mice. Male Balb/c mice (20-25 gm, 26 per group) were treated with an LD75 dose of LPS (15 mg/kg) injected intraperitoneally (IP). Anti-HMGB 1 B box or non-immune rabbit serum (0.3 ml per mouse each time, IP) was given at time 0, +12 hours and +24 hours after LPS administration. Survival of mice was evaluated over time. The results of this study are shown in FIG. 10B, which is a graph ofthe survival of septic mice administered anti-HMGB 1 B box antibodies or non-immune serum. As shown in FIG. 10B, anti-HMGB 1 B box antibodies improved survival ofthe septic mice.
Example 14: Inhibition of HMGBl Signaling Pathway Using an Anti-RAGE Antibody
Previous data implicated RAGE as an HMGBl receptor that can mediate neurite outgrowth during brain development and migration of smooth muscle cells in wound healing (Hori et al. J. Biol. chem. 270:25752-25761, 1995; Merenmies et al. J. Biol. Chem. 266:16722-16729, 1991; and Degryse et al., J. Cell Biol. 152:1197- 1206, 2001). We measured TNF release in RAW 264.7 cultures stimulated with HMGBl (1 μg/ml), LPS (0.1 μg/ml), or HMGBl B box (1 μg/ml) in the presence of anti-RAGE antibody (25 μg/ml) or non-immune IgG (25 μg/ml). Briefly, the cells were seeded into 24-well tissue culture plates and used at 90% confluence. LPS (E. coli 0111:B4, Sigma, St. Louis, MO) was sonicated for 20 minutes before use. Cells were treated with HMGBl (HMG-1; 1 μg/ml), LPS (0.1 μg/ml), or HMGBl B box (B Box; 1 μg/ml) in the presence of anti-RAGΕ antibody (25 μg/ml) or non-immune IgG (25 μg/ml), as indicated in FIG. 11 A, for 16 hours in serum-free Opti-MΕM I medium (Life Technologies) and supematants were collected for TNF measurement using the L929 cytotoxicity assay described herein. IgG purified polyclonal anti- RAGΕ antibody (Catalog No. sc-8230, N-16, Santa Cruz Biotech, Inc., Santa Cruz, CA) was dialyzed extensively against PBS before use. The results of this study are shown in FIG. 11 A, which is a histogram ofthe effects of HMGB 1, LPS, or HMGB 1 B box in the presence of anti-RAGΕ antibodies or non-immune IgG (control) on TNF release from RAW 264.7 cells. As shown in FIG. 11 A, compared to non-immune IgG, anti-RAGΕ antibody significantly inhibited HMGBl B box-induced TNF release. This suppression was specific, because anti-RAGΕ did not significantly inhibit LPS-stimulated TNF release. Notably, the maximum inhibitory effect of anti- RAGE decreased HMG-1 signaling by only 40%, suggesting that other signal transduction pathways may participate in HMGBl signaling.
To examine the effects of HMGB 1 or HMGB 1 B box on the NF-κB-dependent ELAM promoter, the following experiment was carried out. RAW 264.7 macrophages were transiently co-transfected with an expression plasmid encoding a murine MyD 88-dominant-negative (DN) mutant (corresponding to amino acids 146-296), or empty vector, plus a luciferase reporter plasmid under the control ofthe NF-κB -dependent ELAM promoter, as described by Means et al. (J. Immunol. 166:4074-4082, 2001). A portion ofthe cells were then stimulated with full-length HMGBl (100 ng/ml), or purified HMGBl B box (10 μg/ml), for 5 hours. Cells were then harvested and luciferase activity was measured, using standard methods. All transfections were performed in triplicate, repeated at least three times, and a single representative experiment is shown in FIG. 1 IB. As shown in FIG. 1 IB, HMGBl stimulated luciferase activity in samples that were not co-transfected with the MyD 88 dominant negative, and the level of stimulation was decreased in samples that were co-transfected with the MyD 88 dominant negative. This effect was also observed in samples administered HMGB B box.
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope ofthe invention encompassed by the appended claims.

Claims

CLAIMSWhat is claimed is:
1. A polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, wherein said HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1-395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMGlL8 A box.
2. A polypeptide comprising a high mobility group box protein (HMGB) A box which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, wherein said HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1-
395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box.
3. A polypeptide wherein the polypeptide is a high mobility group box protein (HMGB) A box biologically active fragment or variant thereof which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, wherein said HMGB A box biologically active fragment is selected from the group consisting of an HMG1L5 A box fragment, an HMG1L1 A box fragment, an HMG1L4 A box fragment, an HMGB A box polypeptide of BAC clone RPl 1-395A23 fragment, an HMG1L9 A box fragment, an LOC122441 A box fragment, an LOC139603
A box fragment, and an HMG1L8 A box fragment.
4. A polypeptide wherein the polypeptide is a high mobility group box protein (HMGB) A box biologically active fragment which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein, wherein said HMGB A box biologically active fragment is selected from the group consisting of an HMG1L5 A box fragment, an
HMG1L1 A box fragment, an HMG1L4 A box fragment, an HMGB A box polypeptide fragment of BAC clone RPl 1-395A23, an HMG1L9 A box fragment, an LOCI 22441 A box fragment, an LOCI 39603 A box fragment, and an HMG1L8 A box fragment.
5. A composition comprising a polypeptide comprising a high mobility box protein (HMGB) A box or variant thereof which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in a pharmaceutically acceptable carrier, wherein said HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of
BAC clone RPl 1-395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box.
6. A composition comprising a polypeptide comprising a high mobility box protein (HMGB) A box which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in a pharmaceutically acceptable carrier, wherein said HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMG1L1 A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1-395A23, an HMG1L9 A box, an LOC122441 A box, an LOCI 39603 A box, and an HMG1L8 A box.
7. A composition comprising a polypeptide wherein the polypeptide is a high mobility group box protein (HMGB) A box biologically active fragment or variant thereof which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in a pharmaceutically acceptable carrier, wherein said HMGB A box biologically active fragment is selected from the group consisting of an HMG1L5 A box fragment, an HMG1L1 A box fragment, an HMG1L4 A box fragment, an HMGB A box polypeptide fragment of BAC clone RPl 1-395A23, an
HMG1L9 A box fragment, an LOC122441 A box fragment, an LOC139603 A box fragment, and an HMG1L8 A box fragment.
8. A composition comprising a polypeptide wherein the polypeptide is a high mobility group box protein (HMGB) A box biologically active fragment which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in a pharmaceutically acceptable carrier, wherein said HMGB A box biologically active fragment is selected from the group consisting of an HMG1L5 A box fragment, an HMG1L1 A box fragment, an HMG1L4 A box fragment, an HMGB A box polypeptide fragment of BAC clone RPl 1-395A23, an HMG1L9 A box fragment, an LOCI 22441 A box fragment, an LOCI 39603 A box fragment, and an HMG1L8 A box fragment.
9. A purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, wherein said antibodies can inhibit release of a proinflammatory cytokine from a cell treated with HMGB, wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMG1L1 B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
10. A polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, but not comprising a full length HMGB, wherein said polypeptide can cause release of a proinflammatory cytokine from a cell, and wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23.
11. A polypeptide comprising a high mobility group box protein (HMGB) B box, but not comprising a full length HMGB, wherein said polypeptide can cause release of a proinflammatory cytokine from a cell, and wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23.
12. A polypeptide wherein the polypeptide is a high mobility group box protein (HMGB) B box biologically active fragment or variant thereof, wherein said
HMGB B box biologically active fragment is selected from the group consisting of an HMG1L5 B box fragment, an HMGILI B box fragment, an HMG1L4 B box fragment, and an HMGB B box polypeptide fragment of BAC clone RPl 1-395A23.
13. A polypeptide wherein the polypeptide is a high mobility group box protein (HMGB) B box biologically active fragment, wherein said HMGB B box biologically active fragment is selected from the group consisting of an HMG1L5 B box fragment, an HMGILI B box fragment, an HMG1L4 B box fragment, and an HMGB B box polypeptide fragment of BAC clone RPl 1- 395A23.
14. A method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically bind to non-B box epitopes of HMGB, in an amount sufficient to inhibit the inflammatory cytokine cascade, wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
15. A method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a polypeptide comprising a high mobility group box protein (HMGB) A box or variant thereof which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in an amount sufficient to inhibit release ofthe proinflammatory cytokine from the cell, wherein said HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMGILI A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1-395A23, an HMG1L9 A box, an LOC122441 B box, an LOC139603 A box, and an HMG1L8 A box.
16. A method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade, comprising administering to the patient a polypeptide, wherein said polypeptide is a high mobility group box protein
(HMGB) A box biologically active fragment or variant thereof which can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein in an amount sufficient to inhibit release ofthe proinflammatory cytokine from the cell, wherein said HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMGILI A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1- 395A23 A box, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMG1L8 A box.
17. A method for effecting weight loss or treating obesity in a patient, comprising administering to the patient an effective amount of a polypeptide comprising a high mobility group box protein (HMGB) B box or variant thereof, but not comprising a full length HMGB polypeptide, in an amount sufficient to stimulate the release of a proinflammatory cytokine from a cell, wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
18. A method for effecting weight loss or treating obesity in a patient, comprising administering to the patient an effective amount of a polypeptide, wherein said polypeptide is a high mobility group box protein (HMGB) B box biologically active fragment or a variant thereof in an amount sufficient to stimulate the release of a proinflammatory cytokine from a cell, wherein said HMGB B box biologically active fragment is selected from the group consisting of an HMG1L5 B box fragment, an HMGILI B box fragment, an
HMG1L4 B box fragment, and an HMGB B box polypeptide fragment of BAC clone RPl 1-395A23 B box.
19. A method of determining whether a compound inhibits inflammation, comprising combining the compound with (a) a cell that releases a proinflammatory cytokine when exposed to a high mobility group box protein (HMGB) B box or a biologically active fragment thereof; and (b) the HMGB B box or biologically active fragment thereof, wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an
HMGB B box polypeptide of BAC clone RPl 1-395A23; and then determining whether the compound inhibits the release ofthe proinflammatory cytokine from the cell.
20. A pharmaceutical composition comprising a polypeptide comprising a high mobility group box (HMGB) A box or a fragment or variant thereof that can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box (HMGB) protein and an agent that inhibits TNF biological activity, said agent selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide, in a pharmaceutically acceptable carrier.
21. The pharmaceutical composition of Claim 20, wherein said polypeptide is a mammalian HMGB A box.
22. The pharmaceutical composition of Claim 21 , wherein said polypeptide is a mammalian HMGBl A box.
23. The pharmaceutical composition of Claim 22, wherein said polypeptide comprises SEQ ID NO:4.
24. The pharmaceutical composition of Claim 23, wherein said polypeptide consists of SEQ ID NO:4.
25. The pharmaceutical composition of Claim 21 , wherein said mammalian HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMGILI A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1-395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an HMGlL8 A box.
26. A pharmaceutical composition comprising an antibody that binds an HMGB polypeptide or a biologically active fragment thereof and an agent that inhibits TNF biological activity, said agent selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide, in a pharmaceutically acceptable carrier.
27. The pharmaceutical composition of Claim 26, wherein said HMGB polypeptide is a mammalian HMGB polypeptide.
28. The pharmaceutical composition of Claim 27, wherein said HMGB polypeptide is an HMGBl polypeptide.
29. The pharmaceutical composition of Claim 28, wherein said HMGB 1 polypeptide comprises SEQ ID NO:l.
30. The pharmaceutical composition of Claim 29, wherein said HMGB 1 polypeptide consists of SEQ ID NO: 1.
31. The pharmaceutical composition of Claim 26, wherein said biologically active HMGB fragment is an HMGB B box or a biologically active fragment thereof.
32. The pharmaceutical composition of Claim 31 , wherein said HMGB B box consists of SEQ ID NO:5.
33. The pharmaceutical composition of Claim 31 , wherein said HMGB B box biologically active fragment consists of SEQ ID NO:23.
34. The pharmaceutical composition of Claim 31 , wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RP11-395A23.
35. The pharmaceutical composition of Claim 26, wherein said antibody is a monoclonal antibody.
36. The pharmaceutical composition of Claim 26, wherein said antibody is a polyclonal antibody.
37. A method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade comprising administering to said patient a composition comprising a polypeptide comprising a high mobility group box (HMGB) A box or a fragment or variant thereof that can inhibit release of a proinflammatory cytokine from a cell treated with high mobility group box
(HMGB) protein and an agent that inhibits TNF biological activity, said agent selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide.
38. The method of Claim 37, wherein said composition further comprises a pharmaceutically acceptable carrier.
39. The method of Claim 37, wherein said polypeptide is a mammalian HMGB A box.
40. The method of Claim 39, wherein said polypeptide is a mammalian HMGB 1 A box.
41. The method of Claim 40, wherein said polypeptide comprises SEQ ID NO :4.
42. The method of Claim 41, wherein said polypeptide consists of SEQ ID NO:4.
43. The method of Claim 39, wherein said mammalian HMGB A box is selected from the group consisting of an HMG1L5 A box, an HMGILI A box, an HMG1L4 A box, an HMGB A box polypeptide of BAC clone RPl 1-395A23, an HMG1L9 A box, an LOC122441 A box, an LOC139603 A box, and an
HMGlL8 A box.
44. The method of Claim 37, wherein said condition is selected from the group consisting of sepsis, allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstmctive pulmonary disease, psoriasis, pancreatitis, peritonitis, bums, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative colitis, multiple sclerosis, and cachexia.
45. A method of treating a condition in a patient characterized by activation of an inflammatory cytokine cascade comprising administering to said patient a composition comprising an antibody that binds an HMGB polypeptide or a biologically active fragment thereof and an agent that inhibits TNF biological activity, said agent selected from the group consisting of infliximab, etanercept, adalimumab, CDP870, CDP571, Lenercept, and Thalidomide.
46. The method of Claim 45, wherein said composition further comprises a pharmaceutically acceptable carrier.
47. The method of Claim 45, wherein said (HMGB) polypeptide is a mammalian HMGB polypeptide.
48. The method of Claim 47, wherein said HMGB polypeptide is an HMGB 1 polypeptide.
49. The method of Claim 48, wherein said HMGBl polypeptide comprises SEQ ID NO:l.
50. The method of Claim 49, wherein said HMGB 1 polypeptide consists of SEQ ID NO:l.
51. The method of Claim 45, wherein said biologically active HMGB fragment is an HMGB B box or a biologically active fragment thereof.
52. The method of Claim 51 , wherein said HMGB B box is selected from the group consisting of an HMG1L5 B box, an HMGILI B box, an HMG1L4 B box, and an HMGB B box polypeptide of BAC clone RPl 1-395A23.
53. The method of Claim 51 , wherein said HMGB 1 B box consists of SEQ ID NO:5.
54. The method of Claim 51 , wherein said HMGB 1 B box biologically active fragment consists of SEQ ID NO:23.
55. The method of Claim 45, wherein said antibody is a monoclonal antibody.
56. The method of Claim 45, wherein said antibody is a polyclonal antibody.
57. The method of Claim 45, wherein said condition is selected from the group consisting of sepsis, allograft rejection, rheumatoid arthritis, asthma, lupus, adult respiratory distress syndrome, chronic obstructive pulmonary disease, psoriasis, pancreatitis, peritonitis, burns, myocardial ischemia, organic ischemia, reperfusion ischemia, Behcet's disease, graft versus host disease, Crohn's disease, ulcerative colitis, multiple sclerosis, and cachexia.
PCT/US2003/037507 2002-11-20 2003-11-20 Use of hmgb fragments as anti-inflammatory agents WO2004046345A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA002506328A CA2506328A1 (en) 2002-11-20 2003-11-20 Use of hmgb fragments as anti-inflammatory agents
AU2003294488A AU2003294488B2 (en) 2002-11-20 2003-11-20 Use of HMGB fragments as anti-inflammatory agents
NZ540067A NZ540067A (en) 2002-11-20 2003-11-20 A purified preparation of antibodies that specifically bind to a high mobility group box protein (HMGB) B box but do not specifically to non-B box epitopes of HMGB
EP03789973A EP1569684A4 (en) 2002-11-20 2003-11-20 Use of hmgb fragments as anti-inflammatory agents
JP2004554020A JP2006510619A (en) 2002-11-20 2003-11-20 Use of HMGB fragments as anti-inflammatory agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US42784602P 2002-11-20 2002-11-20
US42784102P 2002-11-20 2002-11-20
US60/427,841 2002-11-20
US60/427,846 2002-11-20

Publications (2)

Publication Number Publication Date
WO2004046345A2 true WO2004046345A2 (en) 2004-06-03
WO2004046345A3 WO2004046345A3 (en) 2004-12-02

Family

ID=32329191

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/037507 WO2004046345A2 (en) 2002-11-20 2003-11-20 Use of hmgb fragments as anti-inflammatory agents

Country Status (6)

Country Link
EP (1) EP1569684A4 (en)
JP (1) JP2006510619A (en)
AU (1) AU2003294488B2 (en)
CA (1) CA2506328A1 (en)
NZ (1) NZ540067A (en)
WO (1) WO2004046345A2 (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005025604A2 (en) * 2003-09-10 2005-03-24 The General Hospital Corporation Use of hmgb and hmgb fragments to decrease specific immune response
WO2006002971A2 (en) * 2004-07-02 2006-01-12 Creabilis Therapeutics S.P.A. Nucleic acids for the treatment of hmgb1-related pathologies
WO2006012373A2 (en) * 2004-07-20 2006-02-02 Critical Therapeutics, Inc. Combination therapies of hmgb and complement inhibitors against inflammation
WO2006024547A3 (en) * 2004-09-03 2006-06-01 Creabilis Therapeutics Spa Protease resistant human and non-human hmgb1 box-a mutants and their therapeutic/diagnostic use
WO2007031100A1 (en) * 2005-09-14 2007-03-22 Ostini, Marco Active immunotherapy of life-threatening systemic inflammation
WO2007054090A1 (en) * 2005-11-09 2007-05-18 Pharmexa A/S Therapeutic vaccines targeting hmgb1
WO2006138429A3 (en) * 2005-06-16 2007-06-21 The Feinstein Inst Medical Res Antibodies against hmgb1 and fragments thereof
WO2007011606A3 (en) * 2005-07-18 2007-07-12 Critical Therapeutics Inc USE OF HMGBl ANTAGONISTS FOR THE TREATMENT OF INFLAMMATORY SKIN CONDITIONS
JP2008520552A (en) * 2004-10-22 2008-06-19 メディミューン,インコーポレーテッド High affinity antibody against HMGB1 and method of use thereof
US7632500B2 (en) 2003-09-11 2009-12-15 Cornerstone Therapeutics, Inc. Monoclonal antibodies against HMGB1
US7749959B2 (en) 2001-05-15 2010-07-06 The Feinstein Institute For Medical Research Use of HMGB fragments as anti-inflammatory agents
US8053206B2 (en) 1999-02-11 2011-11-08 The Feinstein Institute For Medical Research Antagonists of HMG1 for treating inflammatory conditions
US8188041B2 (en) 2003-06-06 2012-05-29 The Feinstein Institute For Medical Research Inhibitors of the interaction between HMGB polypeptides and toll-like receptor 2 as anti-inflammatory agents
US8470325B2 (en) 2007-02-15 2013-06-25 Kagoshima University Method of treating amykloidosis comprising administering an anti-HMGB-1 antibody
US8546547B2 (en) 2006-09-15 2013-10-01 Creabilis Therapeutics S.P.A. Polymer conjugates of Box-A of HMGB1 and Box-A variants of HMGB1
US8604178B2 (en) 2006-09-18 2013-12-10 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses
WO2014115430A1 (en) 2013-01-28 2014-07-31 株式会社イーベック Humanized anti-hmgb1 antibody or antigen-binding fragment thereof
US8956618B2 (en) 2010-01-21 2015-02-17 The Texas A&M University System Vaccine vectors and methods of enhancing immune responses
US8961990B2 (en) 2010-06-09 2015-02-24 The Board Of Trustees Of The University Of Arkansas Vaccine and methods to reduce campylobacter infection
KR20150102957A (en) * 2012-10-25 2015-09-09 가부시키가이샤 제노믹스 Novel method for treating spinal cord injury using HMGB1 fragment
US9603915B2 (en) 2013-02-14 2017-03-28 The Board of Trustees of the University of Akansas Compositions and methods of enhancing immune responses to Eimeria or limiting Eimeria infection
US9623078B2 (en) 2012-10-25 2017-04-18 Genomix Co., Ltd. Method for treating cardiac infarction using HMGB1 fragment
US9919010B2 (en) 2008-04-30 2018-03-20 Genomix Co., Ltd. Method for collecting functional cells in vivo with high efficiency
US10364276B2 (en) 2011-04-26 2019-07-30 StemRIM Inc. Peptide for inducing regeneration of tissue and use thereof
US10376571B2 (en) 2013-03-15 2019-08-13 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to enteric pathogens
US10682398B2 (en) 2016-05-03 2020-06-16 The Texas A&M University System Yeast vaccine vector including immunostimulatory and antigenic polypeptides and methods of using the same
CN111542335A (en) * 2017-12-01 2020-08-14 斯特姆里姆有限公司 Therapeutic agent for inflammatory bowel disease
CN111743890A (en) * 2019-03-26 2020-10-09 深圳先进技术研究院 Application of daminomycin or derivatives thereof
CN112823038A (en) * 2018-10-05 2021-05-18 国家儿童医院研究所 HMGB1 protein derivatives for removal of biofilm
US11191786B2 (en) 2009-10-28 2021-12-07 StemRIM Inc. Agents for promoting tissue regeneration by recruiting bone marrow mesenchymal stem cells and/or pluripotent stem cells into blood
EP3750553A4 (en) * 2018-02-08 2022-01-12 Stemrim Inc. Therapeutic agent for psoriasis
US11746136B2 (en) 2017-03-15 2023-09-05 Research Institute At Nationwide Children's Hospital Composition and methods for disruption of bacterial biofilms without accompanying inflammation
US11969459B2 (en) 2017-01-27 2024-04-30 StemRIM Inc. Therapeutic agent for cardiomyopathy, old myocardial infarction and chronic heart failure

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6303321B1 (en) * 1999-02-11 2001-10-16 North Shore-Long Island Jewish Research Institute Methods for diagnosing sepsis
CA2296792A1 (en) * 1999-02-26 2000-08-26 Genset S.A. Expressed sequence tags and encoded human proteins

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ABRAHAM ET AL.: 'Cutting edge: HMG-1 as a mediator of acute lung inflammation' J. IMMUNOL. vol. 165, no. 6, 15 September 2000, pages 2950 - 2954, XP002982921 *
ANDERSSON ET AL.: 'High mobility group 1 protein (HMG-1) stimulates proinföammatory cytokine synthesis in human monocytes' J. EXP. MED. vol. 192, no. 4, 21 August 2000, pages 565 - 570, XP002233473 *
See also references of EP1569684A2 *
WANG ET AL.: 'HMG-1 as a late mediator of endotoxin lethality in mice' SCIENCE vol. 285, no. 5425, 09 July 1999, pages 248 - 251, XP002940163 *

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8822169B2 (en) 1999-02-11 2014-09-02 The Feinstein Institute For Medical Research HMG1 antibody for treating inflammatory conditions
US8138141B2 (en) 1999-02-11 2012-03-20 The Feinstein Institute For Medical Research HMG1 antibody for treating inflammatory conditions
US8053206B2 (en) 1999-02-11 2011-11-08 The Feinstein Institute For Medical Research Antagonists of HMG1 for treating inflammatory conditions
US7749959B2 (en) 2001-05-15 2010-07-06 The Feinstein Institute For Medical Research Use of HMGB fragments as anti-inflammatory agents
US8501173B2 (en) 2001-05-15 2013-08-06 The General Hospital Corporation Antibodies to high mobility group-1(HMGB1) B-box polypeptides
US7897569B2 (en) 2001-05-15 2011-03-01 The Feinstein Institute For Medical Research Use of HMGB fragments as anti-inflammatory agents
US8188041B2 (en) 2003-06-06 2012-05-29 The Feinstein Institute For Medical Research Inhibitors of the interaction between HMGB polypeptides and toll-like receptor 2 as anti-inflammatory agents
WO2005025604A3 (en) * 2003-09-10 2005-06-09 Gen Hospital Corp Use of hmgb and hmgb fragments to decrease specific immune response
WO2005025604A2 (en) * 2003-09-10 2005-03-24 The General Hospital Corporation Use of hmgb and hmgb fragments to decrease specific immune response
US8846047B2 (en) 2003-09-11 2014-09-30 The Feinstein Institute For Medical Research Monoclonal antibodies against HMGB1
US7632500B2 (en) 2003-09-11 2009-12-15 Cornerstone Therapeutics, Inc. Monoclonal antibodies against HMGB1
WO2006002971A2 (en) * 2004-07-02 2006-01-12 Creabilis Therapeutics S.P.A. Nucleic acids for the treatment of hmgb1-related pathologies
WO2006002971A3 (en) * 2004-07-02 2006-04-20 Creabilis Therapeutics Spa Nucleic acids for the treatment of hmgb1-related pathologies
WO2006012373A2 (en) * 2004-07-20 2006-02-02 Critical Therapeutics, Inc. Combination therapies of hmgb and complement inhibitors against inflammation
WO2006012373A3 (en) * 2004-07-20 2006-04-13 Critical Therapeutics Inc Combination therapies of hmgb and complement inhibitors against inflammation
JP2008511300A (en) * 2004-09-03 2008-04-17 クレアビリス・セラピューティクス・エスピーエー Protease resistant human and non-human HMGB1Box-A variants and their use in therapy / diagnosis
US7635679B2 (en) 2004-09-03 2009-12-22 Creabilis Therapeutics S.P.A. Protease resistant mutant of human HMGB1 high affinity binding domain Box-A (HMGB1 Box-A)
WO2006024547A3 (en) * 2004-09-03 2006-06-01 Creabilis Therapeutics Spa Protease resistant human and non-human hmgb1 box-a mutants and their therapeutic/diagnostic use
KR101249287B1 (en) * 2004-09-03 2013-04-01 크리어빌리스 쎄라퓨틱스 에스.피.에이. Protease resistant human and non­human HMGB1 BOX ­A mutants and their therapeutic/diagnostic use
US8058232B2 (en) 2004-09-03 2011-11-15 Creabilis Therapeutics S.P.A. HMGB1 high affinity binding domain Box-A mutants
AU2005279308B2 (en) * 2004-09-03 2012-05-03 Creabilis Therapeutics S.R.L. Protease resistant human and non-human HMGB1 Box-A mutants and their therapeutic/diagnostic use
JP2008520552A (en) * 2004-10-22 2008-06-19 メディミューン,インコーポレーテッド High affinity antibody against HMGB1 and method of use thereof
WO2006138429A3 (en) * 2005-06-16 2007-06-21 The Feinstein Inst Medical Res Antibodies against hmgb1 and fragments thereof
EP2364998A1 (en) * 2005-06-16 2011-09-14 The Feinstein Institute for Medical Research Antibodies against HMGB1 and fragments thereof
US8354106B2 (en) 2005-06-16 2013-01-15 The Feinstein Institute For Medical Research Antibodies against HMGB1 and fragments thereof
WO2007011606A3 (en) * 2005-07-18 2007-07-12 Critical Therapeutics Inc USE OF HMGBl ANTAGONISTS FOR THE TREATMENT OF INFLAMMATORY SKIN CONDITIONS
WO2007031100A1 (en) * 2005-09-14 2007-03-22 Ostini, Marco Active immunotherapy of life-threatening systemic inflammation
WO2007054090A1 (en) * 2005-11-09 2007-05-18 Pharmexa A/S Therapeutic vaccines targeting hmgb1
US8546547B2 (en) 2006-09-15 2013-10-01 Creabilis Therapeutics S.P.A. Polymer conjugates of Box-A of HMGB1 and Box-A variants of HMGB1
US9707298B2 (en) 2006-09-15 2017-07-18 Creabilis Therapeutics S.R.L. Polymer conjugates of Box-A of HMGB1 and Box-A variants of HMGB1
US8604178B2 (en) 2006-09-18 2013-12-10 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses
US9226957B2 (en) 2006-09-18 2016-01-05 The Texas A&M University System Compositions and methods of enhancing immune responses
US10004798B2 (en) 2006-09-18 2018-06-26 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses
US8470325B2 (en) 2007-02-15 2013-06-25 Kagoshima University Method of treating amykloidosis comprising administering an anti-HMGB-1 antibody
US11197895B2 (en) 2008-04-30 2021-12-14 StemRIM Inc. Method for collecting functional cells in vivo with high efficiency
US9919010B2 (en) 2008-04-30 2018-03-20 Genomix Co., Ltd. Method for collecting functional cells in vivo with high efficiency
US11191786B2 (en) 2009-10-28 2021-12-07 StemRIM Inc. Agents for promoting tissue regeneration by recruiting bone marrow mesenchymal stem cells and/or pluripotent stem cells into blood
US8956618B2 (en) 2010-01-21 2015-02-17 The Texas A&M University System Vaccine vectors and methods of enhancing immune responses
US9913893B2 (en) 2010-01-21 2018-03-13 The Board Of Trustees Of The University Of Arkansas Vaccine vectors and methods of enhancing immune responses
US8961990B2 (en) 2010-06-09 2015-02-24 The Board Of Trustees Of The University Of Arkansas Vaccine and methods to reduce campylobacter infection
US10960068B2 (en) 2010-06-09 2021-03-30 The Board Of Trustees Of The University Of Arkansas Vaccine and methods to reduce campylobacter infection
US10550165B2 (en) 2011-04-26 2020-02-04 StemRIM Inc. Peptide for inducing regeneration of tissue and use thereof
US10364276B2 (en) 2011-04-26 2019-07-30 StemRIM Inc. Peptide for inducing regeneration of tissue and use thereof
US9623078B2 (en) 2012-10-25 2017-04-18 Genomix Co., Ltd. Method for treating cardiac infarction using HMGB1 fragment
AU2013335685B2 (en) * 2012-10-25 2017-10-12 Osaka University Novel method for treating spinal cord injury using HMGB1 fragment
US9688733B2 (en) 2012-10-25 2017-06-27 Genomix Co., Ltd. Method for treating spinal cord injury using HMGB1 fragment
KR20150102957A (en) * 2012-10-25 2015-09-09 가부시키가이샤 제노믹스 Novel method for treating spinal cord injury using HMGB1 fragment
EP2913059A4 (en) * 2012-10-25 2016-03-30 Genomix Co Ltd Novel method for treating spinal cord injury using hmgb1 fragment
KR102146822B1 (en) * 2012-10-25 2020-08-21 가부시키가이샤 스템림 Novel method for treating spinal cord injury using HMGB1 fragment
WO2014115430A1 (en) 2013-01-28 2014-07-31 株式会社イーベック Humanized anti-hmgb1 antibody or antigen-binding fragment thereof
US9550825B2 (en) 2013-01-28 2017-01-24 Evec Inc. Humanized anti-HMGB1 antibody or antigen-binding fragment thereof
US9603915B2 (en) 2013-02-14 2017-03-28 The Board of Trustees of the University of Akansas Compositions and methods of enhancing immune responses to Eimeria or limiting Eimeria infection
US9884099B2 (en) 2013-02-14 2018-02-06 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to Eimeria or limiting Eimeria infection
US10328137B2 (en) 2013-02-14 2019-06-25 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to Eimeria or limiting Eimeria infection
US10792351B2 (en) 2013-02-14 2020-10-06 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to Eimeria or limiting Eimeria infection
US11904005B2 (en) 2013-02-14 2024-02-20 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to Eimeria or limiting Eimeria infection
US11364290B2 (en) 2013-02-14 2022-06-21 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to eimeria or limiting eimeria infection
US11013792B2 (en) 2013-03-15 2021-05-25 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to enteric pathogens
US10376571B2 (en) 2013-03-15 2019-08-13 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to enteric pathogens
US10716840B2 (en) 2013-03-15 2020-07-21 The Board Of Trustees Of The University Of Arkansas Compositions and methods of enhancing immune responses to enteric pathogens
US10682398B2 (en) 2016-05-03 2020-06-16 The Texas A&M University System Yeast vaccine vector including immunostimulatory and antigenic polypeptides and methods of using the same
US11382962B2 (en) 2016-05-03 2022-07-12 The Board Of Trustees Of The University Of Arkansas Yeast vaccine vector including immunostimulatory and antigenic polypeptides and methods of using the same
US11969459B2 (en) 2017-01-27 2024-04-30 StemRIM Inc. Therapeutic agent for cardiomyopathy, old myocardial infarction and chronic heart failure
US11746136B2 (en) 2017-03-15 2023-09-05 Research Institute At Nationwide Children's Hospital Composition and methods for disruption of bacterial biofilms without accompanying inflammation
US11298403B2 (en) 2017-12-01 2022-04-12 StemRIM Inc. Therapeutic agent for inflammatory bowel disease
CN111542335A (en) * 2017-12-01 2020-08-14 斯特姆里姆有限公司 Therapeutic agent for inflammatory bowel disease
EP3750553A4 (en) * 2018-02-08 2022-01-12 Stemrim Inc. Therapeutic agent for psoriasis
EP3860718A4 (en) * 2018-10-05 2022-08-31 Research Institute at Nationwide Children's Hospital Hmgb1 protein derivatives for the removal of biofilms cross-reference to related application
CN112823038A (en) * 2018-10-05 2021-05-18 国家儿童医院研究所 HMGB1 protein derivatives for removal of biofilm
CN111743890A (en) * 2019-03-26 2020-10-09 深圳先进技术研究院 Application of daminomycin or derivatives thereof

Also Published As

Publication number Publication date
WO2004046345A3 (en) 2004-12-02
EP1569684A2 (en) 2005-09-07
NZ540067A (en) 2007-05-31
JP2006510619A (en) 2006-03-30
CA2506328A1 (en) 2004-06-03
AU2003294488A1 (en) 2004-06-15
AU2003294488B2 (en) 2007-05-24
EP1569684A4 (en) 2006-08-02

Similar Documents

Publication Publication Date Title
AU2003294488B2 (en) Use of HMGB fragments as anti-inflammatory agents
US8501173B2 (en) Antibodies to high mobility group-1(HMGB1) B-box polypeptides
AU2002309829B2 (en) Use of HMG fragment as anti-inflammatory agents
US20080124320A1 (en) Use of HMGB fragments as anti-inflammatory agents
AU2002309829A1 (en) Use of HMG fragment as anti-inflammatory agents
JP2005512507A6 (en) Use of HMG fragments as anti-inflammatory agents
US20060111287A1 (en) Acetylated protein
EA003675B1 (en) Interleukin-18 binding proteins, their preparation and use
EP1668035A2 (en) Monoclonal antibodies against hmgb1
JP2008500013A (en) TIM-3 polypeptide
US20040156851A1 (en) HMGB1 combination therapies
JP2003532370A (en) Novel Th2-specific molecules and methods of using same
US5905146A (en) DNA binding protein S1-3
KR20100080769A (en) Modulators of hypersensitivity reactions
AU2007205777A1 (en) Use of HMGB fragments as anti-inflammatory agents
KR20010085816A (en) Compositions and Methods for the Treatment of Immune Related Diseases
AU2007234583B2 (en) Use of HMG fragment as anti-inflammatory agents
JP2003000271A (en) New polypeptide, new dna, new antibody and new gene- modified animal

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003294488

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2506328

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 540067

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2004554020

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003789973

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003789973

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2003294488

Country of ref document: AU