US20050226865A1 - Methods and compositions for treating and preventing diseases associated with alphavbeta5 integrin - Google Patents

Methods and compositions for treating and preventing diseases associated with alphavbeta5 integrin Download PDF

Info

Publication number
US20050226865A1
US20050226865A1 US11/095,945 US9594505A US2005226865A1 US 20050226865 A1 US20050226865 A1 US 20050226865A1 US 9594505 A US9594505 A US 9594505A US 2005226865 A1 US2005226865 A1 US 2005226865A1
Authority
US
United States
Prior art keywords
antibody
alkyl
alula
αvβ5
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/095,945
Other languages
English (en)
Inventor
Dean Sheppard
Amha Atakilit
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
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 University of California filed Critical University of California
Priority to US11/095,945 priority Critical patent/US20050226865A1/en
Assigned to REGENTS OF THE UNIVERSITY OF CALIFORNIA, THE reassignment REGENTS OF THE UNIVERSITY OF CALIFORNIA, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ATAKILIT, AMHA, SHEPPARD, DEAN
Publication of US20050226865A1 publication Critical patent/US20050226865A1/en
Priority to US12/505,366 priority patent/US7815908B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen

Definitions

  • Pulmonary edema (“PE”) affects millions of people each year, causing substantial morbidity and mortality.
  • PE patients the alveoli flood with liquid from pulmonary capillaries which compromises oxygen transfer to the systemic circulation (Hall, et al. in C URRENT T HERAPY IN R ESPIRATORY M EDICINE (R. Cherniack, Ed., 1986), pp. 222-227). This sequence of events results in hypoxemia, hypercapnia, and death if no corrective measures are taken.
  • PE cardiovascular disease
  • ARDS Acute Lung Injury/Adult (acute) Respiratory Distress Syndrome or “ARDS,” which can develop as a result of lung injury due to, e.g., pneumonia, septic shock, trauma, aspiration of vomit, or chemical inhalation, is often associated with non-cardiogenic PE.
  • Non-cardiogenic PE is characterized by a change in the vascular permeability of the lung tissue which leads to an increase in fluid levels in the lungs.
  • Cardiogenic PE is often caused by left sided heart failure and can be a complication of a heart attack, leaking or narrowed heart valves (mitral or aortic valves), or any disease of the heart that either results in weakening and/or stiffening of the heart muscle (cardiomyopathy).
  • the failing heart transmits its increased pressure to the lung veins. As pressure in the lung veins rises, fluid is pushed into the air spaces (alveoli). This fluid then becomes a barrier to normal oxygen exchange, resulting in shortness of breath.
  • Cardiogenic PE is characterized by increased capillary hydrostatic pressure which leads to an increase in fluid levels in the lungs.
  • PE is caused by, e.g., altered capillary permeability; infection; inhaled or circulating toxins; vasoactive substances (e.g., histamine, kinins); disseminated intravascular coagulation; immunologic reactions; radiation-associated pneumonia; uremia; near-drowning; smoke inhalation; and acute respiratory distress syndrome; left ventricular failure; mitral stenosis; bacterial endocarditis; pulmonary venous fibrosis; congenital stenosis of the origin of the pulmonary veins; pulmonary venoocclusive disease; overinfusion of fluids; hypoalbuminemia (e.g., from renal, hepatic, nutritional, or protein-losing enteropathy); high-altitude; drug overdoses, CNS trauma, subarachnoid bleeding, pulmonary embolism, pulmonary parenchymal disease, eclampsia, anesthesia, and cardiopulmonary bypass operations.
  • vasoactive substances e.g., histamine
  • Symptoms of PE may include, for example, shortness of breath, rapid and/or labored breathing, tachycardia, hypertension, tightness in the chest, cold extremities with or without accompanying cyanosis, cough with a frothy or pink sputum, extensive use of accessory muscles of respiration, moist rales with or without wheezing, and combinations thereof.
  • Tests to diagnose PE include blood tests such as complete blood count (CBC), blood urea nitrogen (BUN), creatinine, and serum protein.
  • Urianalysis, arterial blood gases (ABGs), chest X-rays, and electrocardiograms (ECG) and all used to assist the physician in narrowing the diagnosis down to PE.
  • Treatment of of cardiogenic PE typically involves placing the patient on 100% oxygen, morphine to ease anxiety and provide some beneficial cardiac effects, furosemide for diuresis, vasodilators to reduce the work against which the myocardium must pump, and inotropic drugs such as doputamine to increase cardiac contractility.
  • Other measures that have been used are rotating tourniquets on three of four limbs and reducing blood volume by 500 ml.
  • the present invention provides compositions and methods for treating or preventing diseases involving ⁇ v ⁇ 5 integrin such as PE.
  • One embodiment of the present invention provides methods of treating or preventing PE in a mammalian subject (e.g., a primate such as a human, a monkey, or a chimpanzee; a canine; or a feline).
  • a therapeutic amount or prophylactic amount of an antagonist of ⁇ v ⁇ 5 integrin is administered to the subject.
  • the antagonist may be, for example, an agent under 1 kDa, under 0.5 KDa, or under 0.25 KDa.
  • the antagonist may be an antibody or an antibody fragment, including humanized antibodies, scFv, Fab, or (Fab′)2.
  • the antibody may be ALULA, humanized ALULA, or may compete with ALULA for specific binding to ⁇ v ⁇ 5 integrin.
  • Administration may be, but is not limited to intravenous, intra-nasal, or intra-bronchial.
  • the methods of the invention are useful for treating individuals that have PE or are at risk of developing PE.
  • a second therapeutic agent for treating or preventing acute lung injury and/or ARDS and or PE may be administered to the subject, including, but not limited to, e.g., TGF ⁇ pathway inhibitors, activated Protein C, steroids, GM-CSF, diuretic agents, bronchodilating agents, platelet inhibitors, an antibody that binds to ⁇ v ⁇ 5 integrin, an antibody that binds to ⁇ 5, a second antagonist of ⁇ v ⁇ 5 integrin, an antagonist of ⁇ v ⁇ 6 integrin, a ⁇ 2 agonist, or a surfactant.
  • Another embodiment of the invention provides an antibody that specifically competes with ALULA for binding to ⁇ v ⁇ 5 integrin.
  • the antibody of this embodiment may be ALULA itself, humanized ALULA, a fragment of ALULA including, e.g., a scFv, a Fab, and a (Fab′)2 of ALULA, or another antibody that competes with ALULA for binding to ⁇ v ⁇ 5 integrin.
  • the invention also provides pharmaceutical compositions comprising such antibodies and a pharmaceutically acceptable excipient.
  • the pharmaceutical compositions may further comprise a second therapeutic agent (e.g., a TGF ⁇ pathway inhibitor, activated Protein C, a steroid, GM-CSF, a platelet inhibitor, a diuretic agent; a bronchodilating agent, an antibody that binds to ⁇ v ⁇ 5 integrin, an antibody that binds to ⁇ 5, a second antagonist of ⁇ v ⁇ 5 integrin, and an antagonist of ⁇ v ⁇ 6 integrin.) that treats or prevents PE.
  • a second therapeutic agent e.g., a TGF ⁇ pathway inhibitor, activated Protein C, a steroid, GM-CSF, a platelet inhibitor, a diuretic agent; a bronchodilating agent, an antibody that binds to ⁇ v ⁇ 5 integrin, an antibody that binds to ⁇ 5, a second antagonist of ⁇ v ⁇ 5 integrin, and an antagonist of ⁇ v ⁇ 6 integrin.
  • the pharmaceutical compositions may comprise a therapeutic agent for
  • a further embodiment of the invention provides methods of identifying an agent for treating PE.
  • the methods comprise contacting a plurality of agents with ⁇ v ⁇ 5 integrin, selecting an agent that competes with binding of a ligand to ⁇ v ⁇ 5 integrin, and determining the effect of the selected agent on PE.
  • Agents which have an effect on PE are identified as agents for treating PE.
  • the plurality of agents may be a plurality of antibodies or may be under 1 KDa.
  • the ligand may be an antibody, including, e.g., ALULA, or may be vitronectin, fibronectin, osteopontin, tenascin c and adenovirus penton base.
  • kits for treating or preventing PE comprise an antagonist of ⁇ v ⁇ 5 integrin (e.g., a monoclonal antibody including ALULA, and antibodies that compete with ALULA for binding to ⁇ v ⁇ 5 integrin) and a second therapeutic agent for treating pulmonary edema (e.g., a TGF ⁇ pathway inhibitor, activated Protein C, a steroid, GM-CSF, a platelet inhibitor, a diuretic agent; a bronchodilating agent, an antibody that binds to ⁇ v ⁇ 5 integrin, an antibody that binds to ⁇ 5, a second antagonist of ⁇ v ⁇ 5 integrin, an antagonist of ⁇ v ⁇ 6 integrin, a ⁇ 2 agonist, or a surfactant.
  • a TGF ⁇ pathway inhibitor e.g., activated Protein C, a steroid, GM-CSF, a platelet inhibitor, a diuretic agent
  • a bronchodilating agent e.g.,
  • FIG. 1 illustrates results from in vivo experiments that demonstrate that 05/mice are protected from lung injury induced PE.
  • FIG. 2 illustrates results from in vivo experiments that demonstrate that an antibody that specifically binds to ⁇ 5 (i.e., ALULA) reduces the severity of ischemia reperfusion induced PE.
  • ALULA an antibody that specifically binds to ⁇ 5
  • FIG. 3 illustrates results from in vivo experiments that demonstrate that an antibody that specifically binds to ⁇ 5 (i.e., ALULA) reduces the severity of induced by lung injury from large tidal volume ventilation.
  • ALULA antibody that specifically binds to ⁇ 5
  • FIG. 4 illustrates results from in vitro experiments that demonstrate that an antibody that specifically binds to ⁇ 5 (i.e., ALULA) blocks adhesion of cells expressing ⁇ v ⁇ 5 integrin to dishes coated with a range of concentrations of the ⁇ v ⁇ 5 integrin ligand, vitronectin.
  • ALULA antibody that specifically binds to ⁇ 5
  • the present invention is based in part on the surprising discovery that treating animals with agents that bind to ⁇ v ⁇ 5 integrins reduces symptoms of PE. More particularly, blocking binding of ligands to ⁇ v ⁇ 5 integrin can reduce the severity of PE.
  • the inventors have demonstrated that an antibody that binds to ⁇ v ⁇ 5 integrin blocks binding of vitronectin, a ligand of ⁇ v ⁇ 5 integrin, to ⁇ v ⁇ 5 integrin.
  • administration of an antibody that binds to ⁇ v ⁇ 5 integrin reduces the severity of PE. Accordingly, the invention provides methods of treating or preventing PE in a subject by administering an effective amount of an antagonist of ⁇ v ⁇ 5 to the subject.
  • the invention also provides antibodies that compete with the disclosed antibody designated “ALULA” as well as pharmaceutical compositions comprising such antibodies.
  • ALULA binds to ⁇ v ⁇ 5 integrin and administration of ALULA to a mammalian subject reduces the severity of PE in the subject.
  • the invention also provides methods of identifying new agents for the treatment of PE by identifying agents that interact with ⁇ v ⁇ 5 integrins and testing them for their ability to treat PE.
  • ⁇ v ⁇ 5 antagonist is any agent that competes with an ⁇ v ⁇ 5 ligand for available ligand binding sites on ⁇ v ⁇ 5 integrins.
  • ⁇ v ⁇ 5 antagonists include agents that specifically bind to ⁇ v ⁇ 5, ⁇ 5, as well as agent that bind to ⁇ v ⁇ 5 or ⁇ 5 and at least one other integrin such as, e.g., ⁇ v ⁇ 3 or ⁇ v ⁇ 6.
  • ⁇ v ⁇ 5 integrin is a member of a family of adhesion molecules that comprise non-covalently associated ⁇ / ⁇ heterodimers that mediate, inter alia, cell-cell interactions, cell-extracellular matrix interactions, and cell-pathogen interactions.
  • ⁇ v ⁇ 5 is the only integrin that contains the ⁇ 5 subunit.
  • ⁇ v ⁇ 5 recognizes the RGD peptide sequence and binds vitronectin (see, e.g., Hynes, Cell 69:11-25 (1992) and has been implicated in multiple disorders including stroke, myocardial infarction, cancer (i.e., angiogenesis), and ocular neovascularization disease (see, e.g., Friedlander et al., Science 270(5241):1500-2 (1995); Friedlander et al., PNAS USA 93(18):9764-9 (1996); Elicieri et al., J. Cell Biol. 157(10:149-159 (2002); Heba et al., J. Vasc. Res.
  • a “therapeutic dose” or “therapeutically effective amount” or “effective amount” of an ⁇ v ⁇ 5 integrin antagonist is an amount of the antagonist which prevents, alleviates, abates, or reduces the severity of symptoms of diseases associated with ⁇ v ⁇ 5 integrin including, e.g., stroke, myocardial infarction, cancer (i.e., angiogenesis), ocular neovascularization disease, and PE (e.g., fluid accumulation in the lungs, increased pulmonary capillary hydrostatic pressure, or shortness of breath) in a patient.
  • cancer i.e., angiogenesis
  • PE e.g., fluid accumulation in the lungs, increased pulmonary capillary hydrostatic pressure, or shortness of breath
  • antibody refers to a polypeptide encoded by an immunoglobulin gene or functional fragments thereof that specifically binds and recognizes an antigen.
  • the recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon, and mu constant region genes, as well as the myriad immunoglobulin variable region genes.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
  • An exemplary immunoglobulin (antibody) structural unit comprises a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • variable heavy chain refers to the variable region of an immunoglobulin heavy chain, including an Fv, scFv, dsFv or Fab
  • variable light chain refers to the variable region of an immunoglobulin light chain, including of an Fv, scFv, dsFv or Fab.
  • antibody functional fragments include, but are not limited to, complete antibody molecules, antibody fragments, such as Fv, single chain Fv (scFv), complementarity determining regions (CDRs), VL (light chain variable region), VH (heavy chain variable region), Fab, F(ab)2′ and any combination of those or any other functional portion of an immunoglobulin peptide capable of binding to target antigen (see, e.g., F UNDAMENTAL I MMUNOLOGY (Paul ed., 4th ed. 2001).
  • various antibody fragments can be obtained by a variety of methods, for example, digestion of an intact antibody with an enzyme, such as pepsin; or de novo synthesis.
  • Antibody fragments are often synthesized de novo either chemically or by using recombinant DNA methodology.
  • the term antibody includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al., (1990) Nature 348:552).
  • the term “antibody” also includes bivalent or bispecific molecules, diabodies, triabodies, and tetrabodies. Bivalent and bispecific molecules are described in, e.g., Kostelny et al. (1992) J. Immunol.
  • a “humanized” antibody is an antibody that retains the reactivity of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody with their human counterparts. See, e.g., Morrison et al., PNAS USA, 81:6851-6855 (1984); Morrison and Oi, Adv. Immunol., 44:65-92 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988); Padlan, Molec. Immun., 28:489-498 (1991); Padlan, Molec. Immun., 31(3):169-217 (1994).
  • Single chain Fv (scFv) or “single chain antibodies” refers to a protein wherein the V H and the V L regions of a scFv antibody comprise a single chain which is folded to create an antigen binding site similar to that found in two chain antibodies. Methods of making scFv antibodies have been described in e.g., Ward et al., Exp Hematol . (5):660-4 (1993); and Vaughan et al., Nat Biotechnol. 14(3):309-14 (1996).
  • Single chain Fv (scFv) antibodies optionally include a peptide linker of no more than 50 amino acids, generally no more than 40 amino acids, preferably no more than 30 amino acids, and more preferably no more than 20 amino acids in length.
  • the peptide linker is a concatamer of the sequence Gly-Gly-Gly-Gly-Ser, e.g., 2, 3, 4, 5, or 6 such sequences.
  • Gly-Gly-Gly-Gly-Gly-Ser e.g., 2, 3, 4, 5, or 6 such sequences.
  • a valine can be substituted for a glycine.
  • Additional peptide linkers and their use are well-known in the art. See, e.g., Huston et al., Proc.
  • the specified antibodies bind to a particular protein (e.g., ⁇ v ⁇ 5 integrin, ⁇ 5, or portions thereof) and do not bind in a significant amount to other proteins present in the sample.
  • Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein.
  • antibodies raised against an ⁇ v ⁇ 5 integrins or a ⁇ 5 polypeptide can be selected to obtain antibodies specifically immunoreactive with that protein and not with other proteins, except for polymorphic variants, e.g., proteins at least 80%, 85%, 90%, 95% or 99% identical to a sequence of interest.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays, Western blots, or immunohistochemistry are routinely used to select monoclonal antibodies specifically immunoreactive with a protein.
  • a specific or selective reaction will be at least twice the background signal or noise and more typically more than 10 to 100 times background.
  • An agent that “specifically competes” for binding reduces the specific binding of an antibody to a polypeptide.
  • a first antibody is considered to competitively inhibit binding of a second antibody, if binding of the second antibody to the antigen is reduced by at least 30%, usually at least about 40%, 50%, 60% or 75%, and often by at least about 90%, in the presence of the first antibody using any of the competitive binding assays known in the art (see, e.g., Harlow and Lane, supra).
  • polypeptide “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues.
  • the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.
  • the terms encompass amino acid chains of any length, including full length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, ⁇ -carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an ⁇ carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • peptidomimetic and “mimetic” refer to a synthetic chemical compound that has substantially the same structural and functional characteristics of the ⁇ v ⁇ 5 antagonists of the invention.
  • Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed “peptide mimetics” or “peptidomimetics” (see, e.g., Fauchere, J. Adv. Drug Res. 15:29 (1986); Veber and Freidinger TINS p. 392 (1985); and Evans et al. J. Med. Chem. 30:1229 (1987)).
  • Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent or enhanced therapeutic or prophylactic effect.
  • peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biological or pharmacological activity), such as a naturally occurring ⁇ v ⁇ 5 ligand, but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of, e.g., —CH2NH—, —CH2S—, —CH2-CH2-, —CH ⁇ CH— (cis and trans), —COCH2-, —CH(OH)CH2-, and —CH2SO—.
  • a paradigm polypeptide i.e., a polypeptide that has a biological or pharmacological activity
  • a linkage selected from the group consisting of, e.g., —CH2NH—, —CH2S—, —CH2-CH2-, —CH
  • the mimetic can be either entirely composed of synthetic, non-natural analogues of amino acids, or, is a chimeric molecule of partly natural peptide amino acids and partly non-natural analogs of amino acids.
  • the mimetic can also incorporate any amount of natural amino acid conservative substitutions as long as such substitutions also do not substantially alter the mimetic's structure and/or activity.
  • nucleic acid and “polynucleotide” are used interchangeably.
  • polynucleotide includes oligonucleotides (i.e., short polynucleotides).
  • This term also refers to deoxyribonucleotides, ribonucleotides, and naturally occurring variants, and can also refer to synthetic and/or non-naturally occurring nucleic acids (i.e., comprising nucleic acid analogues or modified backbone residues or linkages), such as, for example and without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs), and the like.
  • nucleic acids i.e., comprising nucleic acid analogues or modified backbone residues or linkages
  • PNAs peptide-nucleic acids
  • nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated.
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (see, e.g., Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Cassol et al. (1992); Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).
  • RNAi refers to a nucleic acid that forms a double stranded RNA, which double stranded RNA has the ability to reduce or inhibit expression of a gene or target gene when the siRNA expressed in the same cell as the gene or target gene (see, e.g., Bass, Nature, 411, 428-429 (2001); Elbashir et al., Nature, 411, 494-498 (2001); WO 00/44895; WO 01/36646; WO 99/32619; WO 00/01846; WO 01/29058; WO 99/07409; and WO 00/44914).
  • siRNA thus refers to the double stranded RNA formed by the complementary strands.
  • the complementary portions of the siRNA that hybridize to form the double stranded molecule typically have substantial or complete identity.
  • an siRNA refers to a nucleic acid that has substantial or complete identity to a target gene and forms a double stranded siRNA.
  • the sequence of the siRNA can correspond to the full length target gene, or a subsequence thereof.
  • the siRNA is at least about 15-50 nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is 15-50 nucleotides in length, and the double stranded siRNA is about 15-50 base pairs in length, preferably about preferably about 20-30 base nucleotides, preferably about 20-25 nucleotides in length, e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • “Silencing” or “downregulation” refers to a detectable decrease of transcription and/or translation of a target sequence, i.e., the sequence targeted by the RNAi, or a decrease in the amount or activity of the target sequence or protein in comparison to the normal level that is detected in the absence of the interfering RNA or other nucleic acid sequence.
  • a detectable decrease can be as small as 5% or 10%, or as great as 80%, 90% or 100%. More typically, a detectable decrease ranges from 20%, 30%, 40%, 50%, 60%, or 70%.
  • the present invention provides methods for treating or preventing diseases involving ⁇ v ⁇ 5 integrin such as, e.g., PE, stroke, myocardial infarction, cancer (e.g., by preventing ⁇ v ⁇ 5 integrin-associated angiogenesis), and ocular neovascular diseases (e.g., by preventing ⁇ v ⁇ 5 integrin-associated angiogenesis), by inhibiting binding of ligands to ⁇ v ⁇ 5 integrin.
  • Any method that inhibits ⁇ v ⁇ 5 integrin expression or ligand binding to ⁇ v ⁇ 5 integrin can be used to treat diseases involving ⁇ v ⁇ 5 integrin according to the methods of the invention.
  • antibodies that specifically bind to ⁇ v ⁇ 5 integrin antibodies that specifically bind to the ⁇ 5 subunit, ligands of ⁇ v ⁇ 5 integrin, and peptide, non-peptide, and peptidomimetic analogs of such ligands can be used to inhibit binding to ⁇ v ⁇ 5 integrin and thus, treat or prevent diseases involving ⁇ v ⁇ 5.
  • polynucleotides that inhibit expression of ⁇ 5 e.g., siRNA molecules, antisense sequences, etc.
  • the disease is PE (including, e.g., cardiogenic and non-cardiogenic PE).
  • treatment of PE also treats or prevents downstream disorders such as, e.g., pulmonary fibrosis.
  • antibodies that specifically bind to ⁇ v ⁇ 5 integrin or to the ⁇ 5 subunit of the ⁇ v ⁇ 5 integrin are used to treat or prevent diseases involving ⁇ v ⁇ 5 including, e.g., PE, stroke, myocardial infarction, cancer (i.e., angiogenesis), and ocular neovascular diseases (i.e., angiogenesis).
  • the antibodies may also compete with other ligands for binding to ⁇ v ⁇ 5 integrin or to the ⁇ 5 subunit of the ⁇ v ⁇ 5 integrin.
  • Suitable antibodies include, e.g., monoclonal antibodies, humanized antibodies and antibody fragments (i.e., Fv, Fab, (Fab′) 2 , or scFv).
  • the monoclonal antibody ALULA (ATCC Deposit No. PTA-5817, made Feb. 13, 2004, at the ATCC, 10801 University Boulevard. Manassas, Va. 20110-2209) which binds to the 05 subunit of the ⁇ v ⁇ 5 integrin, is used to treat or prevent diseases involving ⁇ v ⁇ 5 integrin, including, e.g., PE, stroke, myocardial infarction, cancer (i.e., angiogenesis), and ocular neovascular diseases (i.e., angiogenesis).
  • ALULA acts by blocking ⁇ v ⁇ 5 integrin-mediated changes to vascular permeability in the lungs.
  • humanized ALULA, ALULA fragments, or a monoclonal antibody which competes with ALULA for binding to ⁇ v ⁇ 5 integrin or the ⁇ 5 subunit of the ⁇ v ⁇ 5 integrin is used to treat PE.
  • Monoclonal antibodies are obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see, for example, Kohler & Milstein, Eur. J. Immunol. 6: 511-519 (1976)). Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art. Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host. Alternatively, one may isolate DNA sequences which encode a monoclonal antibody or a binding fragment thereof by screening a DNA library from human B cells according to the general protocol outlined by Huse et al., Science 246: 1275-1281 (1989).
  • Monoclonal antibodies are collected and titered against the immunogen in an immunoassay, for example, a solid phase immunoassay with the immunogen immobilized on a solid support.
  • Monoclonal antibodies will usually bind with a K d of at least about 0.1 mM, more usually at least about 1 ⁇ M, preferably at least about 0.1 ⁇ M or better, and most preferably, 0.01 ⁇ M or better.
  • an animal such as a rabbit or mouse is immunized with ⁇ v ⁇ 5 polypeptide, or an nucleic acid construct encoding such a polypeptide.
  • the antibodies produced as a result of the immunization can be isolated using standard methods.
  • the immunoglobulins, including binding fragments and other derivatives thereof, of the present invention may be produced readily by a variety of recombinant DNA techniques, including by expression in transfected cells (e.g., immortalized eukaryotic cells, such as myeloma or hybridoma cells) or in mice, rats, rabbits, or other vertebrate capable of producing antibodies by well known methods.
  • transfected cells e.g., immortalized eukaryotic cells, such as myeloma or hybridoma cells
  • Suitable source cells for the DNA sequences and host cells for immunoglobulin expression and secretion can be obtained from a number of sources, such as the American Type Culture Collection (Catalogue of Cell Lines and Hybridomas, Fifth edition (1985) Rockville, Md.
  • the antibody is a humanized antibody, i.e., an antibody that retains the reactivity of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody with their human counterparts.
  • a humanized antibody i.e., an antibody that retains the reactivity of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody with their human counterparts. See, e.g., Morrison et al., PNAS USA, 81:6851-6855 (1984); Morrison and Oi, Adv. Immunol., 44:65-92 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988); Padlan, Molec. Immun., 28:489-498 (1991); Padlan, Molec. Immun., 31(3):169-217 (1994
  • polynucleotides comprising a first sequence coding for humanized immunoglobulin framework regions and a second sequence set coding for the desired immunoglobulin complementarity determining regions can be produced synthetically or by combining appropriate cDNA and genomic DNA segments.
  • Human constant region DNA sequences can be isolated in accordance with well known procedures from a variety of human cells.
  • the CDRs for producing the immunoglobulins of the present invention will be similarly derived from monoclonal antibodies capable of specifically binding to ⁇ v ⁇ 5 integrin (e.g., ALULA or antibodies that compete with ALULA for specific binding to ⁇ v ⁇ 5 integrin).
  • back mutation can be introduced into the framework regions of the human portion of the antibody. Methods of making back mutations are well known in the art and are described in, e.g., Co et al., PNAS USA 88; 2269-2273 (1991) and WO 90/07861.
  • the antibodies are antibody fragments such as Fab, F(ab′) 2 , Fv or scFv.
  • the antibody fragments can be generated using any means known in the art including, chemical digestion (e.g., papain or pepsin) and recombinant methods. Methods for isolating and preparing recombinant nucleic acids are known to those skilled in the art (see, Sambrook et al., Molecular Cloning. A Laboratory Manual (2d ed. 1989); Ausubel et al., Current Protocols in Molecular Biology (1995)).
  • the antibodies can be expressed in a variety of host cells, including E. coli , other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO, and HeLa cells lines and myeloma cell lines.
  • One embodiment of the invention provides methods for identifying antibodies that compete with ALULA for specific binding to ⁇ v ⁇ 5 integrin.
  • ком ⁇ онентs can be used to identify antibodies that compete with ALULA for specific binding to ⁇ v ⁇ 5 integrin. Any of a number of competitive binding assays known in the art can be used to measure competition between two antibodies to the same antigen. Briefly, the ability of different antibodies to inhibit the binding of another antibody is tested. For example, antibodies can be differentiated by the epitope to which they bind using a sandwich ELISA assay. This is carried out by using a capture antibody to coat the surface of a well. A subsaturating concentration of tagged-antigen is then added to the capture surface. This protein will be bound to the antibody through a specific antibody:epitope interaction.
  • a second antibody which has been covalently linked to a detectable moiety (e.g., HRP, with the labeled antibody being defined as the detection antibody) is added to the ELISA. If this antibody recognizes the same epitope as the capture antibody it will be unable to bind to the target protein as that particular epitope will no longer be available for binding. If however this second antibody recognizes a different epitope on the target protein it will be able to bind and this binding can be detected by quantifying the level of activity (and hence antibody bound) using a relevant substrate.
  • the background is defined by using a single antibody as both capture and detection antibody, whereas the maximal signal can be established by capturing with an antigen specific antibody and detecting with an antibody to the tag on the antigen. By using the background and maximal signals as references, antibodies can be assessed in a pair-wise manner to determine epitope specificity.
  • a first antibody is considered to competitively inhibit binding of a second antibody, if binding of the second antibody to the antigen is reduced by at least 30%, usually at least about 40%, 50%, 60% or 75%, and often by at least about 90%, in the presence of the first antibody using any of the assays described above.
  • any small molecule antagonist of ⁇ v ⁇ 5 can be used according to the methods of the invention to treat or prevent PE.
  • the small molecules will be less than 1000 daltons in mass and will often be less than 500 daltons.
  • Exemplary ⁇ v ⁇ 5 small molecule antagonists include, e.g., those described in U.S. Published Patent Application Nos. 2000/40019206, 2004/0019037, 2004/0019035, 2004/0018192, 2004/0010023, 2003/0181440, 2003/0171271, 2003/0139398, 2002/0037889, 2002/0077321, 2002/0072500, U.S. Pat. No. 6,683,051 and Goodman et al., J. Med. Chem. 45(5):1045-51 (2002).
  • the ⁇ v ⁇ 5 antagonists of the invention will comprise the tri-peptide sequence RGD.
  • the design of such molecules as ligands for the integrins is exemplified, for example, in Pierschbacher et al., J. Cell. Biochem. 56:150-154 (1994)); Ruoslahti, Ann. Rev. Cell. Dev. Bio. 12:697-715 (1996); Chorev et al. Biopolymers 37:367-375 (1995)); Pasqualini et al., J. Cell. Biol. 130:1189-1196 (1995)); Smith et al., J. Biol. Chem. 269:32788-32795 (1994); and U.S. Pat. Nos. 5,780,426 and 6,683,051.
  • the present invention is based on the surprising discovery that blocking binding of ligands to ⁇ v ⁇ 5 integrin reduces the severity of PE.
  • a nucleotide sequence that interferes with the specific expression of the ⁇ v ⁇ 5 integrin gene at the transcriptional or translational level can be used to treat or prevent PE.
  • This approach may utilize, for example, siRNA and/or antisense oligonucleotides to block transcription or translation of a specific mutated mRNA, either by inducing degradation of the mRNA with a siRNA or by masking the mRNA with an antisense nucleic acid.
  • Double stranded siRNA that corresponds to the ⁇ gene can be used to silence the transcription and/or translation of ⁇ v ⁇ 5 integrin by inducing degradation of ⁇ 5 mRNA transcripts, and thus treat or prevent PE by preventing expression of ⁇ v ⁇ 5 integrin.
  • the siRNA is typically about 5 to about 100 nucleotides in length, more typically about 10 to about 50 nucleotides in length, most typically about 15 to about 30 nucleotides in length.
  • siRNA molecules and methods of generating them are described in, e.g., Bass, 2001 , Nature, 411, 428-429; Elbashir et al., 2001 , Nature, 411, 494-498; WO 00/44895; WO 01/36646; WO 99/32619; WO 00/01846; WO 01/29058; WO 99/07409; and WO 00/44914.
  • a DNA molecule that transcribes dsRNA or siRNA also provides RNAi.
  • DNA molecules for transcribing dsRNA are disclosed in U.S. Pat. No. 6,573,099, and in U.S. Patent Application Publication Nos.
  • dsRNA oligonucleotides that specifically hybridize to the nucleic acid sequences set forth in Genbank Accession Nos.: AK054968; BF588784; BE208820; BE207859; or BE206567 can be used in the methods of the present invention.
  • a decrease in the severity of PE symptoms in comparison to symptoms detected in the absence of the interfering RNA can be used to monitor the efficacy of the siRNA.
  • Antisense oligonucleotides that specifically hybridize to nucleic acid sequences encoding ⁇ 5 polypeptides can also be used to silence the transcription and/or translation of ⁇ v ⁇ 5 integrin, and thus treat or prevent PE.
  • antisense oligonucleotides that specifically hybridize to the nucleic acid sequences set forth in Genbank Accession Nos.: BF588784; BE208820; BE207859; BE206567; NM — 002213; BC006541; NM — 174679; AF468059; AY434090; NM — 010580; BC058246; XM — 147237; AF022111; AF022110; AF043257; AF043256; and S58644 can be used in the methods of the present invention.
  • a decrease in the severity of PE symptoms in comparison to symptoms detected in the absence of the antisense nucleic acids can be used to monitor the efficacy of the antis
  • Antisense nucleic acids are DNA or RNA molecules that are complementary to at least a portion of a specific mRNA molecule (see, e.g., Weintraub, Scientific American, 262:40 (1990)). Typically, synthetic antisense oligonucleotides are generally between 15 and 25 bases in length. Antisense nucleic acids may comprise naturally occurring nucleotides or modified nucleotides such as, e.g., phosphorothioate, methylphosphonate, and -anomeric sugar-phosphate, backbone-modified nucleotides.
  • the antisense nucleic acids hybridize to the corresponding mRNA, forming a double-stranded molecule.
  • the antisense nucleic acids interfere with the translation of the mRNA, since the cell will not translate a mRNA that is double-stranded.
  • Antisense oligomers of about 15 nucleotides are preferred, since they are easily synthesized and are less likely to cause problems than larger molecules when introduced into the target nucleotide mutant producing cell.
  • the use of antisense methods to inhibit the in vitro translation of genes is well known in the art (Marcus-Sakura, Anal. Biochem., 172:289, (1988)). Less commonly, antisense molecules which bind directly to the DNA may be used.
  • antisense polynucleotides specific for ⁇ v ⁇ 5 integrin genes can be achieved using any means known in the art including, e.g., direct injection, inhalation, or ingestion of the polynucleotides.
  • antisense polynucleotides can be delivered using a recombinant expression vector (e.g., a viral vector based on an adenovirus, a herpes virus, a vaccinia virus, or a retrovirus) or a colloidal dispersion system (e.g., liposomes) as described herein.
  • a recombinant expression vector e.g., a viral vector based on an adenovirus, a herpes virus, a vaccinia virus, or a retrovirus
  • colloidal dispersion system e.g., liposomes
  • Additional antagonists of ⁇ v ⁇ 5 integrin can be readily identified according to methods well known to those of skill in the art.
  • One convenient method for screening for antagonists involves measuring the ability of the potential antagonists to compete for binding of a known ligand of the integrin.
  • vitronectin, fibronectin, osteopontin, tenascin c and adenovirus penton base are known ligands of ⁇ v ⁇ 5 integrin that can be used in competition assays to identify potential antagonists of ⁇ v ⁇ 5 integrin.
  • Other polypeptides comprising the amino acid sequence RGD can also be used in competition assays.
  • monoclonal antibodies and fragments thereof that bind to ⁇ v ⁇ 5 integrin can be used to screen for additional antagonists of ⁇ v ⁇ 5 integrin.
  • ALULA and antibodies that compete with ALULA for binding to ⁇ v ⁇ 5 are used to screen for additional antagonists of ⁇ v ⁇ 5 integrin.
  • Competition assays are well known in the art. Typically, a ligand of ⁇ v ⁇ 5 integrin or an antibody that competes for ligand binding to ⁇ v ⁇ 5 integrin (e.g., ALULA) is labeled so that differences in binding to ⁇ v ⁇ 5 integrin (e.g., in the presence of increasing amount of a potential competing ligand for ⁇ v ⁇ 5 integrin) can be measured.
  • the ligands may be naturally occurring ligands as well as synthetic ligands. Competition assays indicate the affinity of potential competitor antagonists.
  • the screening methods involve screening a plurality of agents to identify an agent that interacts with ⁇ v ⁇ 5, for example, by binding to ⁇ v ⁇ 5 integrin or preventing an antibody (e.g., ALULA) or ligand specific for ⁇ v ⁇ 5 integrin (e.g., vitronectin, fibronectin, osteopontin, tenascin c, adenovirus penton base) from binding to ( ⁇ v ⁇ 5 integrin.
  • an antibody e.g., ALULA
  • ligand specific for ⁇ v ⁇ 5 integrin e.g., vitronectin, fibronectin, osteopontin, tenascin c, adenovirus penton base
  • Preliminary screens can be conducted by screening for agents capable of binding to o ⁇ v ⁇ 5 integrin, as at least some of the agents so identified are likely ⁇ v ⁇ 5 integrin antagonists.
  • the binding assays usually involve contacting ⁇ v ⁇ 5 integrin with one or more test agents and allowing sufficient time for ⁇ v ⁇ 5 integrin and test agents to form a binding complex. Any binding complexes formed can be detected using any of a number of established analytical techniques. Protein binding assays include, but are not limited to, immunohistochemical binding assays, flow cytometry or other assays.
  • the ⁇ v ⁇ 5 integrin utilized in such assays can be naturally expressed, cloned or synthesized.
  • the screening methods of the invention can be performed as in vitro or cell-based assays.
  • Cell based assays can be performed in any cells in which ⁇ v ⁇ 5 integrin is expressed.
  • Cell-based assays may involve whole cells or cell fractions containing ⁇ v ⁇ 5 integrin to screen for agent binding or modulation of ⁇ v ⁇ 5 integrin activity by the agent.
  • ⁇ v ⁇ 5 integrin can be expressed in cells that do not contain endogenous ⁇ v ⁇ 5 integrin. Suitable cell-based assays are described in, e.g., DePaola et al., Annals of Biomedical Engineering 29:1-9 (2001).
  • Agents that are initially identified as interacting with ⁇ v ⁇ 5 integrin can be further tested to validate the apparent activity.
  • Such studies are conducted with suitable cell-based or animal models of PE as described in Example 1 below.
  • the basic format of such methods involves administering a lead compound identified during an initial screen to an animal that serves as a model and then determining if in fact the PE is ameliorated.
  • the animal models utilized in validation studies generally are mammals of any kind. Specific examples of suitable animals include, but are not limited to, primates (e.g., chimpanzees, monkeys, and the like) and rodents (e.g., mice, rats, guinea pigs, rabbits, and the like).
  • the agents tested as potential antagonists of ⁇ v ⁇ 5 integrin can be any small chemical compound, or a biological entity, such as a polypeptide, sugar, nucleic acid or lipid.
  • modulators can be genetically altered versions of ⁇ v ⁇ 5 integrin or an ⁇ v ⁇ 5 integrin ligand.
  • any chemical compound can be used as a potential modulator or ligand in the assays of the invention, although most often compounds that can be dissolved in aqueous or organic (especially DMSO-based) solutions are used.
  • the assays are designed to screen large chemical libraries by automating the assay steps and providing compounds from any convenient source to assays, which are typically run in parallel (e.g., in microtiter formats on microtiter plates in robotic assays).
  • the agents have a molecular weight of less than 1,500 daltons, and in some cases less than 1,000, 800, 600, 500, or 400 daltons.
  • the relatively small size of the agents can be desirable because smaller molecules have a higher likelihood of having physiochemical properties compatible with good pharmacokinetic characteristics, including oral absorption than agents with higher molecular weight.
  • agents less likely to be successful as drugs based on permeability and solubility were described by Lipinski et al.
  • high throughput screening methods involve providing a combinatorial chemical or peptide library containing a large number of potential therapeutic compounds (potential modulator or ligand compounds). Such “combinatorial chemical libraries” or “ligand libraries” are then screened in one or more assays, as described herein, to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional “lead compounds” or can themselves be used as potential or actual therapeutics.
  • a combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis, by combining a number of chemical “building blocks” such as reagents.
  • a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks (amino acids) in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.
  • combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., U.S. Pat. No. 5,010,175, Furka, Int. J. Pept. Prot. Res. 37:487-493 (1991) and Houghton et al., Nature 354:84-88 (1991)).
  • chemistries for generating chemical diversity libraries can also be used. Such chemistries include, but are not limited to: peptoids (e.g., PCT Publication No.
  • nucleic acid libraries see Ausubel, Berger and Sambrook, all supra
  • peptide nucleic acid libraries see, e.g., U.S. Pat. No. 5,539,083
  • antibody libraries see, e.g., Vaughn et al., Nature Biotechnology, 14(3):309-314 (1996) and PCT/US96/10287)
  • carbohydrate libraries see, e.g., Liang et al., Science, 274:1520-1522 (1996) and U.S. Pat. No.
  • compositions comprising antagonists of ⁇ v ⁇ 65 integrin.
  • the compositions of the invention can be provided to treat or prevent diseases which involve ⁇ v ⁇ 5 integrins including, e.g., PE, stroke, myocardial infarction, and cancer (i.e., angiogenesis).
  • compositions of the invention can be provided to treat or prevent PE in subjects with PE or at risk for developing PE.
  • a subject having had exposure to a toxic inhalant would likely be treated after such exposure, whereas a patient at risk of PE can be treated prophylactically and/or therapeutically.
  • patients at risk of PE include patients with acute aspiration, patients exhibiting symptoms of bacterial sepsis, patients whose blood cultures are positive for gram positive or gram negative bacteria, patients with pancreatitis, or patients in hemorrhagic shock.
  • compositions of the invention may be administered on a regular basis (e.g., daily) for a period of time (e.g., 2, 3, 4, 5, 6, days or 1-3 weeks or more).
  • compositions of the invention can be administered directly to the mammalian subject to block ⁇ v ⁇ 5 binding using any route known in the art, including e.g., by injection (e.g., intravenous, intraperitoneal, subcutaneous, intramuscular, or intrademal), inhalation, transdermal application, rectal administration, or oral administration.
  • injection e.g., intravenous, intraperitoneal, subcutaneous, intramuscular, or intrademal
  • inhalation e.g., transdermal application, rectal administration, or oral administration.
  • compositions of the invention may comprise a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there are a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington's Pharmaceutical Sciences, 17th ed., 1989).
  • compositions of the invention can be made into aerosol formulations (i.e., they can be “nebulized”) to be administered via inhalation.
  • Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • Formulations suitable for administration include aqueous and non-aqueous solutions, isotonic sterile solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • compositions can be administered, for example, orally, nasally, topically, intravenously, intraperitoneally, or intrathecally.
  • the formulations of compounds can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials. Solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • the modulators can also be administered as part a of prepared food or drug.
  • Formulations suitable for oral administration can comprise: (a) liquid solutions, such as an effective amount of the packaged nucleic acid suspended in diluents, such as water, saline or PEG 400; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as liquids, solids, granules or gelatin; (c) suspensions in an appropriate liquid; and (d) suitable emulsions.
  • liquid solutions such as an effective amount of the packaged nucleic acid suspended in diluents, such as water, saline or PEG 400
  • capsules, sachets or tablets each containing a predetermined amount of the active ingredient, as liquids, solids, granules or gelatin
  • suspensions in an appropriate liquid such as water, saline or PEG 400
  • Tablet forms can include one or more of lactose, sucrose, mannitol, sorbitol, calcium phosphates, corn starch, potato starch, microcrystalline cellulose, gelatin, colloidal silicon dioxide, talc, magnesium stearate, stearic acid, and other excipients, colorants, fillers, binders, diluents, buffering agents, moistening agents, preservatives, flavoring agents, dyes, disintegrating agents, and pharmaceutically compatible carriers.
  • Lozenge forms can comprise the active ingredient in a flavor, e.g., sucrose, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin or sucrose and acacia emulsions, gels, and the like containing, in addition to the active ingredient, carriers known in the art.
  • a flavor e.g., sucrose
  • an inert base such as gelatin and glycerin or sucrose and acacia emulsions, gels, and the like containing, in addition to the active ingredient, carriers known in the art.
  • the dose administered to a patient should be sufficient to effect a beneficial response in the subject over time, e.g., a reduction in pulmonary capillary hydrostatic pressure, a reduction in fluid in the lungs, a reduction in the rate of fluid accumulation in the lungs, or a combination thereof.
  • the optimal dose level for any patient will depend on a variety of factors including the efficacy of the specific modulator employed, the age, body weight, physical activity, and diet of the patient, on a possible combination with other drugs, and on the severity of the PE.
  • the size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular compound or vector in a particular subject.
  • a physician may evaluate circulating plasma levels of the antagonist and antagonist toxicity.
  • the dose equivalent of an antagonist is from about 1 ng/kg to 10 mg/kg for a typical subject.
  • the antagonists of ⁇ v ⁇ 5 integrin can be administered at a rate determined by the LD 50 of the antagonist, and the side-effects of the antagonist at various concentrations, as applied to the mass and overall health of the subject. Administration can be accomplished via single or divided doses.
  • an antagonist of ⁇ v ⁇ 5 integrin is administered in conjunction with a second therapeutic agent for treating or preventing a disease or disorder associated with ⁇ v ⁇ 5 integrin (e.g., stroke, myocardial infarction, and cancer (i.e., angiogenesis)).
  • a disease or disorder associated with ⁇ v ⁇ 5 integrin e.g., stroke, myocardial infarction, and cancer (i.e., angiogenesis)
  • an antagonist of ⁇ v ⁇ 5 integrin may be administered in conjunction with a second therapeutic agent for treating or preventing acute lung injury and/or ARDS or PE.
  • an antagonist of ⁇ v ⁇ 5 integrin may be administered in conjunction with any of the standard treatments for PE including, e.g., diuretic agents, bronchodilating agents, narcotics, oxygen, and selective tourniquet application.
  • an antagonist of ⁇ v ⁇ 5 integrin may be administered in conjunction with agents that target metabolic pathways that are implicated in acute lung injury and/or ARDS or PE.
  • an antagonist of ⁇ v ⁇ 5 integrin may be administered in conjunction with TGF ⁇ pathway inhibitors, activated Protein C, steroids, GM-CSF, platelet inhibitors, ⁇ -2 agonists, surfactants, antibodies that specifically bind to ⁇ v ⁇ 5 integrin or ⁇ 5, a second antagonist of ⁇ v ⁇ 5 integrin, antibodies that specifically bind to a ⁇ v ⁇ 6 integrin, antagonists of ⁇ v ⁇ 6 integrin, thrombin receptor antagonists, anti-thrombin agents, rho kinase inhibitors, and nucleic acids that inhibit expression of ⁇ v ⁇ 5 integrin including e.g., the antisense oligonucleotides and siRNA described herein.
  • Suitable TGF ⁇ pathway inhibitors include, e.g., TGF- ⁇ antibodies (including those that specifically block TGF- ⁇ 1, TGF- ⁇ 2, TGF- ⁇ 3 or any combination thereof) as described in e.g., Ling et al., J. Amer. Soc. Nephrol. 14: 377-388 (2003), McCormick et al., J. Immunol. 163:5693-5699 (1999), and Cordeiro, Curr. Opin. Mol. Ther. 5(2):199-203 (2003); TGF- ⁇ receptor type II inhibitors or TGF- ⁇ receptor type I kinase inhibitors as described in, e.g., DaCosta Bayfield, Mol. Pharmacol.
  • Suitable ⁇ -2 agonists include, e.g., albuterol, bitolterol, formoterol, isoproterenol, levalbuterol, metaproterenol, pirbuterol, salmeterol, and terbutaline.
  • Suitable surfactants include, e.g., exosurf, infasurf, KL-4, pumactant, survanta, venticute, and surfactant TA, as described in Taeusch et al., Acta Pharmacol Sin 23 Supplement: 11-15 (2002).
  • Suitable anti-thrombin agents include, e.g., hirudin, Hirulog (Biogen), argatroban (Texas Biotechnology) and efegatran (Lilly) and compounds described in U.S. Pat. No. 6,518,244.
  • Suitable thrombin receptor antagonists are described in, e.g., U.S. Pat. Nos.
  • Suitable rho kinase inhibitors include, e.g., Y-27632 as described in e.g., Tasaka et al., Am J Respir Cell Mol. Biol. 2005 Mar. 18; [Epub ahead of print], fasudil as described in, e.g., Nishikimi et al., J Hypertens.
  • the antagonist of ⁇ v ⁇ 5 integrin may be administered combination with an adenovirus expressing ATPase as described in U.S. Patent Publication No. 20020192186; with a ⁇ 2 adrenergic receptor as described in U.S. Patent Publication No. 20020004042; with VEGF ⁇ antagonists as described in U.S. Pat. No. 6,284,751; with lipid peroxidation inhibitors as described in U.S. Pat. No. 5,231,114; and with small molecule inhibitors for ⁇ v ⁇ 6, ⁇ v ⁇ 5, and ⁇ v ⁇ 3 integrins as described in, e.g., U.S. Published Patent Application Nos.
  • the antagonist of ⁇ v ⁇ 5 integrin (e.g., ALULA, humanized ALULA, or fragments of ALULA) and the second therapeutic agent may be administered simultaneously or sequentially.
  • the antagonist of ⁇ v ⁇ 5 integrin may be administered first, followed by the second therapeutic agent.
  • the second therapeutic agent may be administered first, followed by the antagonist of ⁇ v ⁇ 5 integrin.
  • the antagonist of ⁇ v ⁇ 5 integrin and the second therapeutic agent are administered in the same formulation.
  • the antagonist of ⁇ v ⁇ 5 integrin and the second therapeutic agent are administered in different formulations.
  • their administration may be simultaneous or sequential.
  • the antagonists of ⁇ v ⁇ 5 integrin and second therapeutic agent can be administered at a rate determined by the combined LD 50 of the antagonist and the second therapeutic agent, and the side-effects of the antagonist and the second therapeutic agent at various concentrations, as applied to the mass and overall health of the subject.
  • the antagonists of ⁇ v ⁇ 5 integrin and second therapeutic agent are each administered at a subtherapeutic dose or a therapeutic dose.
  • kits for treating or preventing diseases involving ⁇ v ⁇ 5 integrin including, e.g., PE, stroke, myocardial infarction, and cancer (i.e., angiogenesis).
  • the kits comprise an antagonist of ⁇ v ⁇ 5 integrin (e.g., an antibody that binds to ⁇ v ⁇ 5 integrin, including e.g., ALULA, humanized ALULA, or fragments of ALULA), and antibody that binds to ⁇ 5 (e.g., ALULA, humanized ALULA, or fragments of ALULA), and antibody that competes with ALULA) and a second therapeutic agent for treatment of a disease involving ⁇ v ⁇ 5 integrin, including PE.
  • an antagonist of ⁇ v ⁇ 5 integrin e.g., an antibody that binds to ⁇ v ⁇ 5 integrin, including e.g., ALULA, humanized ALULA, or fragments of ALULA
  • antibody that binds to ⁇ 5 e
  • Suitable second therapeutic agents include, e.g., a TGF ⁇ pathway inhibitor, activated Protein C, a steroid, GM-CSF, a platelet inhibitor, a diuretic agent; a bronchodilating agent, antibodies that specifically bind to ⁇ v ⁇ 5 integrin or ⁇ 5, a second antagonist of ⁇ v ⁇ 5 integrin, antibodies that specifically bind to a ⁇ v ⁇ 6 integrin, antagonists of ⁇ v ⁇ 6 integrin, ⁇ -2 agonists, and surfactants.
  • the kits may also comprise written instructions (e.g., a manual) for using the kit.
  • Rodent Single Lung Ischemia-Reperfusion Lung Injury Model of PE Mice or rats undergo lung transplantation, cardiopulmonary bypass, pulmonary thromboendoarterectomy, or severe shock. Next ischemia and reperfusion are induced for thirty minutes and three hours, respectively. To induce ischemia, a left thoracotomy is performed by blocking the left hilum (e.g., with umbilical tape) for 30 minutes. To induce reperfusion, the lungs are reinflated with a tidal volume of 12 ml/kg of air and then normal ventilation is resumed. The animals are euthanized after 3 hours and the permeability of each lung is assessed, e.g., by measuring labeled albumin extravasation into the lung, expressed as extravascular pulmonary equivalents (EVPE).
  • EVPE extravascular pulmonary equivalents
  • Rodent Ventilator-Induced Lung Injury Model of PE Mice or rats are ventilated with normal (6 ml per kg) or high tidal volume (20 ml per kg). Animals are injected with 125 I-labeled albumins after 4 hours and then lungs are harvested and EVPE determined.
  • EVPE Extravascular Plasma Equivalents
  • ALULA was generated as described below.
  • W6/32 a murine monoclonal antibody W6/32 which specifically binds to HLA A, B, and C was obtained from ATCC.
  • CD-1 WT a monoclonal antibody that binds to CD-1 was obtained from ATCC.
  • ALULA a Murine Monoclonal Antibody that Specifically Binds to ⁇ v ⁇ 5 Integrin
  • ⁇ v ⁇ 5 knockout mice were immunized with cells expressing a polypeptide comprising an ⁇ v ⁇ 5 integrin sequence.
  • Monoclonal antibodies that specifically bind ⁇ v ⁇ 5 integrin were identified using methods known in the art. More particularly, ALULA which specifically binds to ⁇ 5 was identified.
  • ALULA was deposited with the ATCC on Feb. 13, 2004 and has the following Accession No.: PTA-5817.
  • ⁇ 5 ⁇ / ⁇ mice and wild type mice were ventilated as described in Example 1 above to induce PE associated with lung injury and EVPE was determined. In contrast to the wild-type mice, ⁇ 5 ⁇ / ⁇ mice did not develop PE after ventilation. These results indicate that ⁇ v ⁇ 5 is involved in PE. The results are shown in FIG. 1 .
  • a Monoclonal Antibody that Specifically Binds to ⁇ 5 Reduces the Severity of Pulmonary Edema Associated with Ischemia Reperfusion
  • rats were given the following treatments and EVPE measurements were taken:
  • Rats that received treatment with ALULA exhibited reduced EVPE (i.e., reduced lung cell permeability) compared to control rats, indicating that a monoclonal antibody that specifically binds to ⁇ 5 can reduce the severity of PE.
  • a Monoclonal Antibody that Specifically Binds to ⁇ 5 Reduces the Severity of Pulmonary Edema Associated With Lung Injury
  • mice were given the following treatments and EVPE measurements were taken:
  • mice that received treatment with ALULA exhibited reduced EVPE compared to control mice, indicating that a monoclonal antibody that specifically binds to ⁇ 5 can reduce the severity of PE.
  • ALULA is the first monoclonal antibody specific for ⁇ v ⁇ 5 that has been shown to have blocking activity in vivo in whole mammals and is the first shown to block increased vascular permeability and the development of alveolar flooding in models of acute lung injury (i.e., PE).
  • SW-480 cells expressing ⁇ v ⁇ 5 integrin are contacted with 0 ⁇ g/ml, 0.1 ⁇ g/ml, 0.3 ⁇ g/ml, and 1 ⁇ g/ml of vitronectin in the presence of 0 ⁇ g/ml, 0.3 ⁇ g/ml, 1 ⁇ g/ml, and 10 ⁇ g/ml of ALULA.
  • a monoclonal antibody i.e., Y9A2 specific for ⁇ 9 ⁇ 1 integrin is used as a negative control.
  • ALULA blocks binding of the ⁇ v ⁇ 5 integrin ligand, vitronectin, to the cells. The results are shown in FIG. 4 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Pulmonology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US11/095,945 2004-04-02 2005-03-30 Methods and compositions for treating and preventing diseases associated with alphavbeta5 integrin Abandoned US20050226865A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/095,945 US20050226865A1 (en) 2004-04-02 2005-03-30 Methods and compositions for treating and preventing diseases associated with alphavbeta5 integrin
US12/505,366 US7815908B2 (en) 2004-04-02 2009-07-17 Methods and compositions for treating and preventing disease associated with αVβ5 integrin

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US55917504P 2004-04-02 2004-04-02
US11/095,945 US20050226865A1 (en) 2004-04-02 2005-03-30 Methods and compositions for treating and preventing diseases associated with alphavbeta5 integrin

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/505,366 Division US7815908B2 (en) 2004-04-02 2009-07-17 Methods and compositions for treating and preventing disease associated with αVβ5 integrin

Publications (1)

Publication Number Publication Date
US20050226865A1 true US20050226865A1 (en) 2005-10-13

Family

ID=35064346

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/095,945 Abandoned US20050226865A1 (en) 2004-04-02 2005-03-30 Methods and compositions for treating and preventing diseases associated with alphavbeta5 integrin
US12/505,366 Active US7815908B2 (en) 2004-04-02 2009-07-17 Methods and compositions for treating and preventing disease associated with αVβ5 integrin

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/505,366 Active US7815908B2 (en) 2004-04-02 2009-07-17 Methods and compositions for treating and preventing disease associated with αVβ5 integrin

Country Status (15)

Country Link
US (2) US20050226865A1 (de)
EP (2) EP2394662B1 (de)
JP (2) JP5223072B2 (de)
CN (1) CN1964740A (de)
AU (1) AU2005229015C1 (de)
CA (1) CA2562038C (de)
CY (1) CY1114497T1 (de)
DK (1) DK1734996T3 (de)
ES (2) ES2671522T3 (de)
HK (1) HK1098077A1 (de)
HR (1) HRP20130782T1 (de)
PL (1) PL1734996T3 (de)
PT (1) PT1734996E (de)
RS (1) RS52918B (de)
WO (1) WO2005094391A2 (de)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011011775A1 (en) 2009-07-24 2011-01-27 The Regents Of The University Of California Methods and compositions for treating and preventing disease associated with avb5 integrin
WO2016040839A1 (en) * 2014-09-12 2016-03-17 Biogen Ma Inc. Humanized anti-alpha v beta 5 antibodies and uses thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9487780B2 (en) * 2012-06-01 2016-11-08 Ionis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with fibronectin
JP2016515120A (ja) * 2013-03-15 2016-05-26 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. 抗アルファvベータ5抗体を用いた急性腎損傷の治療及び予防
WO2014144616A2 (en) * 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 5 antibodies and uses thereof
EP3021942A4 (de) * 2013-07-19 2017-04-19 The Regents of The University of California Epidermaler wachstumsfaktor 8 in form von milchfettkügelchen zur fettsäureaufnahmeregulierung
US11339221B2 (en) 2017-11-01 2022-05-24 Tufts Medical Center, Inc. Bispecific antibody constructs and methods of use

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10023A (en) * 1853-09-20 Machine fob sawing sticks eob broom-handles
US18192A (en) * 1857-09-15 And samuel m
US19037A (en) * 1858-01-05 Washing-machine
US19035A (en) * 1858-01-05 mallary
US19206A (en) * 1858-01-26 Hydrant
US37889A (en) * 1863-03-10 Mode of
US72500A (en) * 1867-12-24 Improvement in bleaceisg and scouring hemp, flax, aid other fibres
US77321A (en) * 1868-04-28 Amos bank
US139398A (en) * 1873-05-27 Improvement in lamp-extinguishers
US171271A (en) * 1875-12-21 Improvement in bob-sleds
US181440A (en) * 1876-08-22 Improvement in steam-boiler furnaces
US5527679A (en) * 1991-05-01 1996-06-18 Dana Farber Cancer Institute β5 protein and DNA encoding the same
US5780426A (en) * 1995-06-07 1998-07-14 Ixsys, Incorporated Fivemer cyclic peptide inhibitors of diseases involving αv β3
US6069158A (en) * 1997-01-08 2000-05-30 Smithkline Beecham Corporation Vitronectin receptor antagonists
US6683051B1 (en) * 1998-09-16 2004-01-27 Merck Patent Gmbh Pharmaceutical preparation containing a cyclopeptide and a chemotherapeutic agent or an angiogenesis inhibitor
US6692741B2 (en) * 1997-08-08 2004-02-17 The Regents Of The University Of California Treatment of acute lung injury and fibrosis with antagonists of β6-specific antibodies
US7053041B1 (en) * 1996-05-31 2006-05-30 The Scripps Research Institute Methods and compositions useful for inhibition of αvβ5mediated angiogenesis

Family Cites Families (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0173494A3 (de) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimäre Rezeptoren durch Verbindung und Expression von DNS
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
JPS63501765A (ja) 1985-11-01 1988-07-21 インタ−ナショナル、ジェネティック、エンジニアリング インコ−ポレ−テッド 抗体遺伝子のモジュ−ル組立体、それにより産生された抗体及び用途
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5010175A (en) 1988-05-02 1991-04-23 The Regents Of The University Of California General method for producing and selecting peptides with specific properties
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
IE66205B1 (en) 1990-06-14 1995-12-13 Paul A Bartlett Polypeptide analogs
US5231114A (en) 1990-06-22 1993-07-27 Wakunaga Seiyaku Kabushiki Kaisha Polysulfides compounds and lipid peroxidation inhibitor containing the polysulfide compounds as active ingredient
US5650489A (en) 1990-07-02 1997-07-22 The Arizona Board Of Regents Random bio-oligomer library, a method of synthesis thereof, and a method of use thereof
DE4123341A1 (de) * 1991-07-15 1993-01-21 Thomae Gmbh Dr K Phenylalkylderivate, diese verbindungen enthaltende arzneimittel und verfahren zu ihrer herstellung
DE4142366A1 (de) * 1991-07-15 1993-06-24 Thomae Gmbh Dr K Phenylalkylderivate, diese verbindungen enthaltende arzneimittel und verfahren zu ihrer herstellung
US5777085A (en) 1991-12-20 1998-07-07 Protein Design Labs, Inc. Humanized antibodies reactive with GPIIB/IIIA
US6291196B1 (en) * 1992-01-31 2001-09-18 Research Corporation Technologies, Inc. Melanoma and prostate cancer specific antibodies for immunodetection and immunotherapy
JPH07507995A (ja) 1992-03-02 1995-09-07 バイオジェン,インコーポレイテッド トロンビンレセプターアンタゴニスト
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US6210671B1 (en) 1992-12-01 2001-04-03 Protein Design Labs, Inc. Humanized antibodies reactive with L-selectin
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5593853A (en) 1994-02-09 1997-01-14 Martek Corporation Generation and screening of synthetic drug libraries
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5525735A (en) 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
US5569588A (en) 1995-08-09 1996-10-29 The Regents Of The University Of California Methods for drug screening
CA2227265C (en) * 1995-08-14 2012-01-03 The Scripps Research Institute Methods and compositions useful for inhibition of .alpha.v.beta.5 mediated angiogenesis
PL329851A1 (en) 1996-05-01 1999-04-12 Lilly Co Eli Treatment of diseases associated with vegf
EP0956865B2 (de) 1996-08-12 2010-08-18 Mitsubishi Tanabe Pharma Corporation MEDIKAMENTE ENTHALTEND Rho-KINASE INHIBITOREN
UA60311C2 (uk) * 1996-10-02 2003-10-15 Смітклайн Бічам Корпорейшн Антагоністи рецептора вітронектину, спосіб одержання цих сполук та фармацевтична композиція
TW589189B (en) 1997-08-04 2004-06-01 Scras Kit containing at least one double-stranded RNA combined with at least one anti-viral agent for therapeutic use in the treatment of a viral disease, notably of viral hepatitis
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
AUPP249298A0 (en) 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
GB9827152D0 (en) 1998-07-03 1999-02-03 Devgen Nv Characterisation of gene function using double stranded rna inhibition
DE69942671D1 (de) 1998-12-01 2010-09-23 Facet Biotech Corp Humanisierte antikoerper gegen gamma-interferon
MXPA01006798A (es) 1998-12-30 2003-06-04 Edema Clearance Inc Terapia de genes para curar edema pulmonar utilizando vectores de adenovirus que codifican para na, k-atppasa.
EP2314700A1 (de) 1999-01-28 2011-04-27 Medical College of Georgia Research Institute, Inc Zusammensetzung und Verfahren zur in vivo und in vitro Abschwächung der Genexpression mittels dobbelsträngiger RNA
DE19956568A1 (de) 1999-01-30 2000-08-17 Roland Kreutzer Verfahren und Medikament zur Hemmung der Expression eines vorgegebenen Gens
US6399581B1 (en) 1999-05-11 2002-06-04 Harbor Branch Oceanographic Institution, Inc. Compound possessing potent thrombin receptor antagonist activity
CN1352641A (zh) 1999-09-29 2002-06-05 奥索-麦克尼尔药品公司 用于治疗整联蛋白介导的疾病的异哌啶甲酰胺
CA2386270A1 (en) 1999-10-15 2001-04-26 University Of Massachusetts Rna interference pathway genes as tools for targeted genetic interference
US6544982B1 (en) 1999-10-29 2003-04-08 Merck & Co., Inc. Thrombin receptor antagonists
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
WO2001053297A1 (en) 2000-01-20 2001-07-26 Merck & Co., Inc. Alpha v integrin receptor antagonists
AU780988B2 (en) 2000-01-24 2005-04-28 Merck & Co., Inc. Alpha V integrin receptor antagonists
US6403612B2 (en) 2000-01-31 2002-06-11 Merck & Co., Inc. Thrombin receptor antagonists
US6515023B2 (en) 2000-01-31 2003-02-04 Merck & Co., Inc. Thrombin receptor antagonists
KR100767000B1 (ko) 2000-02-03 2007-10-15 에자이 알앤드디 매니지먼트 가부시키가이샤 인테그린 발현 저해제
US6518244B2 (en) 2000-03-09 2003-02-11 Intimax Corporation Combinations of heparin cofactor II agonist and platelet IIb/IIIa antagonist, and uses thereof
US6970452B2 (en) 2000-03-13 2005-11-29 Curitell Communications Inc. Common subscriber managing apparatus and method based on functional modeling of a common subscriber server for use in an ALL-IP network and method therefor
AU2001259078A1 (en) 2000-04-18 2001-10-30 Edema Clearance, Inc. Gene theraphy for pulmonary edema using adenovirus vectors encoding a beta2adrenergic receptor gene
AU2001269821A1 (en) 2000-06-15 2001-12-24 Barbara Chen Cycloalkyl alkanoic acids as integrin receptor antagonists
US7288390B2 (en) * 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
DE10041423A1 (de) 2000-08-23 2002-03-07 Merck Patent Gmbh Biphenylderivate
US20020072500A1 (en) 2000-09-27 2002-06-13 Thomas Rogers Hydroxy acid integrin antagonists
DE60118025T2 (de) 2000-10-24 2006-11-23 Merck & Co., Inc. Dibenzoxazepin-alpha-v-integrin-rezeptorantagonist
CA2433680A1 (en) 2000-12-28 2002-08-01 Gregory M Arndt Double-stranded rna-mediated gene suppression
WO2002081627A2 (en) 2001-04-04 2002-10-17 Allergan, Inc. Methods of screening and using inhibitors of angiogenesis
US7081460B2 (en) 2001-04-09 2006-07-25 Ortho-Mcneil Pharmaceutical, Inc. Quinazoline and quinazoline-like compounds for the treatment of integrin-mediated disorders
US20040019206A1 (en) 2001-09-27 2004-01-29 Peter Ruminiski Lactone integrin antagonists
RU2333210C2 (ru) * 2002-08-16 2008-09-10 Янссен Фармацевтика Н.В. Соединения пиперидинила, селективно связывающие интегрины
US20040266755A1 (en) 2003-05-29 2004-12-30 Schering Aktiengesellschaft Prodrugs of 1-(1-hydroxy-5-isoquinolinesulfonyl) homopiperazine
TWI552751B (zh) 2011-06-20 2016-10-11 H 朗德貝克公司 投予4-((1r,3s)-6-氯-3-苯基-二氫茚-1-基)-1,2,2-三甲基-哌及其鹽用於治療精神分裂症的方法

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US139398A (en) * 1873-05-27 Improvement in lamp-extinguishers
US19206A (en) * 1858-01-26 Hydrant
US10023A (en) * 1853-09-20 Machine fob sawing sticks eob broom-handles
US19035A (en) * 1858-01-05 mallary
US171271A (en) * 1875-12-21 Improvement in bob-sleds
US37889A (en) * 1863-03-10 Mode of
US72500A (en) * 1867-12-24 Improvement in bleaceisg and scouring hemp, flax, aid other fibres
US181440A (en) * 1876-08-22 Improvement in steam-boiler furnaces
US19037A (en) * 1858-01-05 Washing-machine
US18192A (en) * 1857-09-15 And samuel m
US77321A (en) * 1868-04-28 Amos bank
US5527679A (en) * 1991-05-01 1996-06-18 Dana Farber Cancer Institute β5 protein and DNA encoding the same
US5780426A (en) * 1995-06-07 1998-07-14 Ixsys, Incorporated Fivemer cyclic peptide inhibitors of diseases involving αv β3
US7053041B1 (en) * 1996-05-31 2006-05-30 The Scripps Research Institute Methods and compositions useful for inhibition of αvβ5mediated angiogenesis
US6069158A (en) * 1997-01-08 2000-05-30 Smithkline Beecham Corporation Vitronectin receptor antagonists
US6692741B2 (en) * 1997-08-08 2004-02-17 The Regents Of The University Of California Treatment of acute lung injury and fibrosis with antagonists of β6-specific antibodies
US6683051B1 (en) * 1998-09-16 2004-01-27 Merck Patent Gmbh Pharmaceutical preparation containing a cyclopeptide and a chemotherapeutic agent or an angiogenesis inhibitor

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011011775A1 (en) 2009-07-24 2011-01-27 The Regents Of The University Of California Methods and compositions for treating and preventing disease associated with avb5 integrin
EP2456460A1 (de) * 2009-07-24 2012-05-30 The Regents of the University of California Verfahren und zusammensetzungen zur behandlung und prävention von mit avb5-integrin assoziierten krankheiten
US20120328604A1 (en) * 2009-07-24 2012-12-27 The Regents Of The University Of California Methods and compositions for treating and preventing disease associated with avb5 integrin
EP2456460A4 (de) * 2009-07-24 2013-02-20 Univ California Verfahren und zusammensetzungen zur behandlung und prävention von mit avb5-integrin assoziierten krankheiten
AU2010275367B2 (en) * 2009-07-24 2015-09-03 The Regents Of The University Of California Methods and compositions for treating and preventing disease associated with avB5 integrin
KR101752515B1 (ko) * 2009-07-24 2017-06-29 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 Avb5 인테그린과 결부된 질환의 치료 및 예방을 위한 방법 및 조성물
US10087252B2 (en) * 2009-07-24 2018-10-02 The Regents Of The University Of California Methods and compositions for treating and preventing disease associated with αvβ5 integrin
WO2016040839A1 (en) * 2014-09-12 2016-03-17 Biogen Ma Inc. Humanized anti-alpha v beta 5 antibodies and uses thereof

Also Published As

Publication number Publication date
WO2005094391A3 (en) 2005-12-01
RS52918B (en) 2014-02-28
JP5681149B2 (ja) 2015-03-04
AU2005229015B2 (en) 2012-08-30
EP2394662A3 (de) 2012-02-22
CA2562038A1 (en) 2005-10-13
EP1734996A4 (de) 2008-09-10
AU2005229015A1 (en) 2005-10-13
ES2671522T3 (es) 2018-06-06
CN1964740A (zh) 2007-05-16
EP2394662B1 (de) 2018-03-21
EP1734996A2 (de) 2006-12-27
US7815908B2 (en) 2010-10-19
AU2005229015C1 (en) 2013-01-17
US20090280118A1 (en) 2009-11-12
JP5223072B2 (ja) 2013-06-26
HRP20130782T1 (en) 2013-09-30
EP1734996B1 (de) 2013-05-22
DK1734996T3 (da) 2013-06-10
HK1098077A1 (en) 2007-07-13
CA2562038C (en) 2017-04-25
JP2007531733A (ja) 2007-11-08
PL1734996T3 (pl) 2013-09-30
JP2013028611A (ja) 2013-02-07
WO2005094391A2 (en) 2005-10-13
EP2394662A2 (de) 2011-12-14
CY1114497T1 (el) 2016-10-05
PT1734996E (pt) 2013-07-18
ES2411965T3 (es) 2013-07-09

Similar Documents

Publication Publication Date Title
US7815908B2 (en) Methods and compositions for treating and preventing disease associated with αVβ5 integrin
DK1734997T3 (en) Natalizumab for use in the treatment of diseases requiring steroid treatment
JP6277127B2 (ja) インスリン様成長因子1による細胞の活性化を阻害するための方法
EP2298348A1 (de) Antikörper gegen Alpha4beta7 Integrin und sein Verwendung zur Behandlung von Darmerkrankungen
CN103272232A (zh) 炎性疾病的预防或治疗药
US10087252B2 (en) Methods and compositions for treating and preventing disease associated with αvβ5 integrin
AU2012216372B2 (en) Methods and compositions for treating and preventing disease associated with alphaVbeta5 integrin
US7875277B2 (en) Compositions and methods for inhibiting squamous cell carcinoma
US20100055114A1 (en) Use of integrin alpha 10 binding antibody to modulate extracellular matrix (cartilage) turnover
AU2011213878B2 (en) Steroid sparing agents and methods of using same
NZ621515B2 (en) Method for inhibiting cellular activation by insulin-like growth factor-1

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENTS OF THE UNIVERSITY OF CALIFORNIA, THE, CALI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SHEPPARD, DEAN;ATAKILIT, AMHA;REEL/FRAME:016426/0287

Effective date: 20050525

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION