WO2009114828A1 - Compositions et procédés relatifs à des états d’allergie gastro-intestinale - Google Patents

Compositions et procédés relatifs à des états d’allergie gastro-intestinale Download PDF

Info

Publication number
WO2009114828A1
WO2009114828A1 PCT/US2009/037192 US2009037192W WO2009114828A1 WO 2009114828 A1 WO2009114828 A1 WO 2009114828A1 US 2009037192 W US2009037192 W US 2009037192W WO 2009114828 A1 WO2009114828 A1 WO 2009114828A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
receptor
ige
antibodies
subject
Prior art date
Application number
PCT/US2009/037192
Other languages
English (en)
Inventor
Marc Rothenberg
Original Assignee
Childrens Hospital Medical Center
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 Childrens Hospital Medical Center filed Critical Childrens Hospital Medical Center
Publication of WO2009114828A1 publication Critical patent/WO2009114828A1/fr

Links

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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons

Definitions

  • Gastrointestinal allergic diseases have dramatically increased in prevalence over the past several decades. Recent studies have found that 2-6% of the population currently suffers from food anaphylaxis. These often debilitating diseases are characterized by a wide array of manifestations including elevated levels of IgE (total and antigen specific), eosinophilia, mastocytosis and gastrointestinal dysfunction (e.g. vomiting, diarrhea and failure-to-thrive).
  • IgE total and antigen specific
  • eosinophilia to mastocytosis
  • gastrointestinal dysfunction e.g. vomiting, diarrhea and failure-to-thrive
  • the present invention provides a method of treating a gastrointestinal allergic condition in a subject, comprising administering to said subject an effective amount of an inhibitor of a T R 2 response.
  • said gastrointestinal allergic condition is intestinal anaphylaxis.
  • said gastrointestinal allergic condition is food anaphylaxis.
  • said subject exhibits at least one symptom selected from the group of symptoms consisting of: elevated levels of total IgE, elevated levels of antigen-specific IgE, elevated levels of a T R 2 cytokine, elevated levels of IL- 4, elevated levels of IL-13, elevated levels of IL-5, elevated levels of IL-9, eosinophilia, mastocytosis, gastrointestinal dysfunction, vomiting, diarrhea and failure-to-thrive.
  • said inhibitor comprises an inhibitor of IL- 4, IL-5, IL-9, IL-13, or IgE.
  • said inhibitor comprises an antibody, antibody fragment, or antibody derivative that specifically binds IL-4, IL-4 receptor, IL- 13, IL- 13 receptor, IL-5, IL-5 receptor, IL-9, IL-9 receptor, IgE, or IgE receptor.
  • said inhibitor comprises a soluble, ligand-binding fragment of a receptor for IL-4, IL-5, IL-9, IL- 13, or IgE.
  • said method comprises the additional step of determining whether said subject has an elevated level of IgE, IL-4, IL-5, IL-9, or IL- 13 before administering said inhibitor.
  • said subject is susceptible to developing said gastrointestinal allergic condition, and said treatment reduces the likelihood that said subject will develop said gastrointestinal allergic condition.
  • said treatment reduces the severity of a symptom of said gastrointestinal allergic condition.
  • said treatment reduces the duration of a symptom of said gastrointestinal allergic condition.
  • the present invention relates to methods and compositions for treating gastrointestinal allergic diseases.
  • Such treatments include, for example, prophylactic methods.
  • prophylactic methods include methods that prevent the onset, or reduce the likelihood of the onset, or reduce the duration or intensity, of one or more symptoms or effects of the gastrointestinal disease.
  • the methods of the invention also include, for example, methods of treating a subject who exhibits one or more symptoms or effects of a gastrointestinal allergic disease.
  • compositions of the invention include, for example, inhibitors of IL-4 or IL- 13 signaling, such as antibodies that bind IL-4, IL- 13, or that bind one of their receptors, soluble receptor fragments, and mutated IL-4 or IL- 13 molecules, as well as inhibitors of IgE and other molecules involved in IL-4 or IL- 13 signaling.
  • inhibitors of IL-4 or IL- 13 signaling such as antibodies that bind IL-4, IL- 13, or that bind one of their receptors, soluble receptor fragments, and mutated IL-4 or IL- 13 molecules, as well as inhibitors of IgE and other molecules involved in IL-4 or IL- 13 signaling.
  • Light chain variable domain (or region),” “heavy chain variable domain (or region),” “CDRl , 2, and 3” and “FRl , 2, 3, and 4" are defined according to the scheme of Kabat et al. in Sequences of Proteins of Immunological Interest, 5 Ed., US Dept. of Health and Human Services, PHS, NIH, NIH Publication no. 91-3242, 1991.
  • a biological molecule e.g. , a polypeptide, antibody, or nucleic acid
  • a biological molecule is "isolated” when it occurs in a location, environment, cell or organism as a result of human intervention (e.g. , in a substantially purified form, in a plasmid or other vector, or in a cell type that does not normally contain or express the molecule).
  • a biological molecule is "substantially purified” if it is sufficiently free of other biological molecules, cell debris, and other substances to be used in standard laboratory protocols (e.g., a binding or hybridization assay). Methods of substantially purifying polypeptides, antibodies, and nucleic acids are well-known in the art.
  • peptide refers to a molecule comprising two or more amino acid residues joined to each other by peptide bonds.
  • nucleic acid molecules e.g., cDNA or genomic DNA
  • RNA molecules e.g., mRNA
  • analogs of the DNA or RNA generated using nucleotide analogs e.g., peptide nucleic acids and non-naturally occurring nucleotide analogs
  • hybrids thereof e.g., peptide nucleic acids and non-naturally occurring nucleotide analogs
  • the nucleic acid molecule can be single-stranded or double-stranded.
  • the nucleic acid molecules of the invention comprise a contiguous open reading frame encoding an antibody, or a fragment, derivative, mutein, or variant thereof, of the invention.
  • a “vector” is a nucleic acid that can be used to introduce another nucleic acid linked to it into a cell.
  • a vector is a "plasmid,” which refers to a linear or circular double stranded DNA molecule into which additional nucleic acid segments can be ligated.
  • a viral vector e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses
  • certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors comprising a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • An "expression vector” is a type of vector that can direct the expression of a chosen polynucleotide.
  • a nucleotide sequence is "operably linked" to a regulatory sequence if the regulatory sequence affects the expression (e.g. , the level, timing, or location of expression) of the nucleotide sequence.
  • a "regulatory sequence” is a nucleic acid that affects the expression (e.g. , the level, timing, or location of expression) of a nucleic acid.
  • the regulatory sequence can, for example, exert its effects directly on the regulated nucleic acid, or through the action of one or more polyeptides (e.g., polypeptides that bind to the regulatory sequence and/or the nucleic acid). Examples of regulatory sequences include promoters, enhancers and other expression control elements (e.g., polyadenylation signals).
  • a "host cell” is a cell that can be used to express a nucleic acid, e.g., a nucleic acid of the invention.
  • a host cell can be a prokaryote, for example, E. coli, or it can be a eukaryote, for example, a single-celled eukaryote ⁇ e.g., a yeast or other fungus), a plant cell ⁇ e.g., a tobacco or tomato plant cell), an animal cell ⁇ e.g., a human cell, a monkey cell, a hamster cell, a rat cell, a mouse cell, or an insect cell) or a hybridoma.
  • host cells examples include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (see Gluzman et al, 1981, Cell 23:175), L cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells or their derivatives such as Veggie CHO and related cell lines which grow in serum-free media (see Rasmussen et al, 1998, Cytotechnology 28:31) or CHO strain DX-Bl 1, which is deficient in DHFR (see Urlaub et al, 1980, Proc. Natl. Acad. Sci.
  • COS-7 line of monkey kidney cells ATCC CRL 1651
  • L cells C127 cells
  • 3T3 cells ATCC CCL 163
  • CHO Chinese hamster ovary
  • HeLa cells derived from the African green monkey kidney cell line CVl (ATCC CCL 70) (see McMahan et al, 1991, EMBO J. 10:2821), human embryonic kidney cells such as 293, 293 EBNA or MSR 293, human epidermal A431 cells, human Colo205 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HL-60, U937, HaK or Jurkat cells.
  • a host cell is a cultured cell that can be transformed or transfected with a polypeptide-encoding nucleic acid, which can then be expressed in the host cell.
  • the phrase "recombinant host cell” can be used to denote a host cell that has been transformed or transfected with a nucleic acid to be expressed.
  • a host cell also can be a cell that comprises the nucleic acid but does not express it at a desired level unless a regulatory sequence is introduced into the host cell such that it becomes operably linked with the nucleic acid. It is understood that the term host cell refers not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a “chimeric antibody” is an antibody in which a portion of the heavy and/or light chain is identical with, homologous to, or derived from an antibody from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is/are identical with, homologous to, or derived from an antibody (-ies) from another species or belonging to another antibody class or subclass. Also included are fragments of such antibodies, that exhibit the desired biological activity (i.e., the ability to specifically bind IL-4 receptor). See, U.S. Patent No. 4,816,567 and Morrison, 1985, Science 229:1202-07.
  • a “CDR grafted antibody” is an antibody comprising one or more CDRs derived from an antibody of a particular species or isotype and the framework of another antibody of the same or different species or isotype.
  • a “multi-specific antibody” is an antibody that recognizes more than one epitope on one or more antigens.
  • a subclass of this type of antibody is a "bi-specific antibody” which recognizes two distinct epitopes on the same or different antigens.
  • a "variant" of a polypeptide comprises an amino acid sequence wherein one or more amino acid residues are inserted into, deleted from and/or substituted into the amino acid sequence relative to another polypeptide sequence.
  • Variants of the invention include fusion proteins.
  • a “derivative" of a polypeptide is a polypeptide (e.g., an antibody) that has been chemically modified in some manner distinct from insertion, deletion, or substitution variants, e.g., via conjugation to another chemical moiety.
  • an antibody includes, in addition to antibodies comprising two full-length heavy chains and two full-length light chains, derivatives, variants, fragments, and muteins thereof, examples of which are described below.
  • An antibody "specifically binds" to its target if it binds to the target immunospecifically.
  • an “antigen binding domain” or “antigen binding region” is the portion of an antibody molecule which contains the amino acid residues (or other moieties) that interact with an antigen and confer on the antibody its specificity and affinity for the antigen.
  • An “epitope” is the portion of a molecule that is bound by an antibody.
  • An epitope can comprise non-contiguous portions of the molecule (e.g., in a polypeptide, amino acid residues that are not contiguous to one another in the polypeptide's sequence but that, together in the context of the molecule, are bound by an antibody).
  • Treatment refers to methods of reducing the severity, duration, or likelihood of onset of a disease or other medical condition, or of a symptom or other effect of a disease or other medical condition, and includes methods of prevention and prophylaxis, as well as attempts to cure, ameliorate, manage or prevent the worsening of a disease or other medical condition.
  • the present invention provides methods of treating allergic conditions, for example, gastrointestinal allergic conditions by inhibiting the activity of the IL-4 or IL- 13 signaling pathways.
  • Such conditions include, for example, intestinal anaphylaxis and food anaphylaxis.
  • Some such conditions are associated with certain symptoms, such as elevated levels of total IgE, elevated levels of antigen- specific IgE, eosinophilia, mastocytosis, gastrointestinal dysfunction, gastrointestinal pain or discomfort, vomiting, diarrhea or failure-to-thrive.
  • the method of treatment comprises reducing the activity of the IL-4 or IL- 13 signaling pathways. The inhibition can occur upstream of, at, or downstream of the IL-4 or IL- 13 receptors using any effective means.
  • inhibition can be achieved through administration of a molecule that binds to IL-4 or IL- 13 (such as a molecule comprising an antibody or a soluble fragment of an IL-4 or IL- 13 receptor), a molecule that binds to an IL-4 receptor or an IL- 13 receptor (such as a molecule comprising an antibody or a mutated IL-4 or IL- 13), or a molecule that interferes with the downstream signal sent by an IL-4 or IL- 13 receptor (such as a small molecule).
  • a molecule that binds to IL-4 or IL- 13 such as a molecule comprising an antibody or a soluble fragment of an IL-4 or IL- 13 receptor
  • a molecule that binds to an IL-4 receptor or an IL- 13 receptor such as a molecule comprising an antibody or a mutated IL-4 or IL- 13
  • a molecule that interferes with the downstream signal sent by an IL-4 or IL- 13 receptor such as
  • the present invention provides antibodies, and fragments, derivatives, muteins, and variants thereof, e.g. , antibodies that bind to human IL-4 receptor alpha.
  • Antibodies that may be employed in accordance with the present invention include antibodies that inhibit a biological activity of IL-4 or IL-13.
  • biological activities include association of receptor component (e.g., IL-2R gamma or IL- 13R alpha), binding (either alone or as part of a multimeric receptor complex) a signaling molecule (e.g., IL-4 or IL-13), and transducing a signal in response to binding a signaling molecule.
  • Different antibodies to the same target may bind to different domains or epitopes of the target or act by different mechanisms of action. Examples include but are not limited to antibodies that interfere with binding of IL-4 to IL-4R or that inhibit signal transduction.
  • the site of action may be, for example, intracellular (e.g. , by interfering with an intracellular signaling cascade) or extracellular.
  • An antibody need not completely inhibit an activity of IL-4, IL- 13, IL-5, IL-9, IgE, etc., to find use in the present invention; rather, antibodies that reduce a particular activity are contemplated for use as well.
  • a compound useful for practicing the instant invention may act by, for example, reducing proliferation, activation, migration, influx, or accumulation of a particular cell type, or by inhibiting a biological response directly or indirectly attributable to a particular cell type.
  • particular cell types are fibroblasts, mast cells, and eosinophils.
  • IL-4 and IL- 13 are associated with a T H 2 response, and are among the cytokines secreted by T R 2 cells.
  • a compound may be administered to reduce a T R 2- type immune response. The compound may be said to reduce proliferation of T H 2 cells, to suppress a T H 2response, to shift the immune response toward a T H 1 response, or to favor a T H l-type response.
  • Such compounds include antagonists of T H 2-type cytokines, such as IL-4, IL-9, IL-5, IL-IO, and IL-13. Techniques for measuring the amount of such cytokines in a subject, e.g., in the subject's serum, are well known.
  • Particular embodiments of methods provided herein comprise administering a compound to inhibit IL-4-induced damage to epithelium in the gastrointestinal tract. Such methods may be employed to prevent epithelial damage, or to restore epithelial barrier function (i.e., promote repair or healing of the epithelium).
  • Combinations of two or more compounds may be employed in the methods and compositions of the present invention.
  • Mutated forms of the various T H 2 -related cytokines, or there receptors, or antibodies or other polypeptides that bind to them, having inhibitory activity can be made using standard techniques. Oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required. Examples of techniques for making such alterations are described in Walder et al, 1986,Gene 42:133; Bauer et al.1985, Gene 37:73; Craik, BioTechniques, January 1985, 12-19; Smith et al, 1981, Genetic Engineering: Principles and Methods, Plenum Press; and U.S. Patent Nos. 4,518,584 and 4,737,462.
  • Oligomers that contain one or more T H 2 cytokine-inhibiting polypeptides may be employed. Oligomers may be in the form of covalently-linked or non-covalently- linked dimers, trimers, or higher oligomers. Oligomers comprising two or more such polypeptides are contemplated for use, with one example being a homodimer. Other oligomers include heterodimers, heterotrimers, etc.
  • One embodiment is directed to oligomers comprising multiple anti-IL-4R antibody polypeptides joined via covalent or non-covalent interactions between peptide moieties fused to the anti-IL-4R antibody polypeptides.
  • Such peptides may be peptide linkers (spacers), or peptides that have the property of promoting oligomerization.
  • Leucine zippers and certain polypeptides derived from antibodies are among the peptides that can promote oligomerization of IL-4R polypeptides attached thereto, as described in more detail below.
  • the oligomers comprise from two to four anti-IL- 4R antibody polypeptides.
  • the anti-IL-4R antibody moieties of the oligomer may be in any of the forms described above, e.g., variants or fragments.
  • the oligomers comprise anti-IL-4R antibody polypeptides that have IL-4R binding activity.
  • an oligomer is prepared using polypeptides derived from immunoglobulins.
  • Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al., 1991, PNAS USA 88:10535; Byrn et al., 1990, Nature 344:677; and Hollenbaugh et al., 1992 "Construction of Immunoglobulin Fusion Proteins", in Current Protocols in Immunology, Suppl. 4, pages 10.19.1 - 10.19.11.
  • Fc polypeptide as used herein includes native and mutein forms of polypeptides derived from the Fc region of an antibody. Truncated forms of such polypeptides containing the hinge region that promotes dimerization are also included. Fusion proteins comprising Fc moieties (and oligomers formed therefrom) offer the advantage of facile purification by affinity chromatography over Protein A or Protein G columns.
  • Fc polypeptide described in PCT application WO 93/10151 (hereby incorporated by reference), is a single chain polypeptide extending from the N-terminal hinge region to the native C-terminus of the Fc region of a human IgGl antibody.
  • Another useful Fc polypeptide is the Fc mutein described in U.S.
  • the oligomer is a fusion protein comprising multiple antibody polypeptides, with or without peptide linkers (spacer peptides).
  • suitable peptide linkers are those described in U.S. Patents 4,751,180 and 4,935,233.
  • Leucine zipper domains are peptides that promote oligomerization of the proteins in which they are found.
  • Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., 1988, Science 240:1759), and have since been found in a variety of different proteins.
  • leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize.
  • leucine zipper domains suitable for producing soluble oligomeric proteins are described in PCT application WO 94/10308, and the leucine zipper derived from lung surfactant protein D (SPD) described in Hoppe et al., 1994, FEBS Letters 344:191, hereby incorporated by reference.
  • SPD lung surfactant protein D
  • the use of a modified leucine zipper that allows for stable trimerization of a heterologous protein fused thereto is described in Fanslow et al., 1994, Semin. Immunol. 6:267-78.
  • recombinant fusion proteins comprising an anti-IL-4R antibody fragment or derivative fused to a leucine zipper peptide are expressed in suitable host cells, and the soluble oligomeric anti-IL-4R antibody fragments or derivatives that form are recovered from the culture supernatant.
  • Antibody-derived polypeptides and fusion proteins described herein may be prepared by any of a number of conventional techniques. For example, they may be purified from cells that naturally express them, or they may be produced in recombinant expression systems, using any technique known in the art.
  • any expression system known in the art can be used to make the recombinant polypeptides of the invention.
  • host cells are transformed with a recombinant expression vector that comprises DNA encoding a desired polypeptide.
  • the host cells that may be employed are prokaryotes, yeast or higher eukaryotic cells.
  • Prokaryotes include gram negative or gram positive organisms, for example E. coli or bacilli.
  • Higher eukaryotic cells include insect cells and established cell lines of mammalian origin.
  • suitable mammalian host cell lines include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (Gluzman et al, 1981, Cell 23:175), L cells, 293 cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells, HeLa cells, BHK (ATCC CRL 10) cell lines, and the CVI/EBNA cell line derived from the African green monkey kidney cell line CVI (ATCC CCL 70) as described by McMahan et al, 1991, EMBO J. 10: 2821.
  • the transformed cells are cultured under conditions that promote expression of the polypeptide, and the polypeptide is recovered by conventional protein purification procedures.
  • One such purification procedure includes the use of affinity chromatography, e.g. , over a matrix having all or a portion (e.g. , the extracellular domain) of the target protein bound thereto.
  • the present invention provides antibodies that interfere with the binding of a T R 2 cytokine to its receptor.
  • Such antibodies referred to herein as blocking antibodies, may be raised against the cytokine or its receptor, or a fragment, variant or derivative thereof, and screened in conventional assays for the ability to interfere with binding of cytokine to receptor.
  • an antibody that blocks binding of IL-4 and also of IL- 13 to cells.
  • the antibodies inhibit IL-4-induced biological activity and also inhibit IL- 13- induced activity, and thus may be employed in treating conditions induced by either or both cytokines.
  • Antibodies that bind to IL-4R alpha may be screened in various conventional assays to determine whether they interfere with the binding of IL- 13 to IL-4R alpha- containing IL- 13 receptor complexes. Antibodies may be screened, for example, in binding assays or tested for the ability to inhibit an IL- IL- 13 -induced biological activity.
  • Antibodies specific for IL-4R alpha may be prepared using any technique known in the art. See, for example, Monoclonal Antibodies, Hybridomas: A New Dimension in Biological Analyses, Kennet et al. (eds.), Plenum Press, New York
  • Antigen-binding fragments of antibodies of the invention may be produced by conventional techniques. Examples of such fragments include, but are not limited to, Fab and F(ab')2 fragments. Antibody fragments and derivatives produced by genetic engineering techniques are also contemplated. Unless otherwise specified, the terms "antibody” and “monoclonal antibody” as used herein encompass both whole antibodies and antigen-binding fragments and/or derivatives thereof.
  • Additional embodiments include chimeric antibodies, e.g., humanized versions of murine monoclonal antibodies.
  • humanized antibodies may be prepared by known techniques, and offer the advantage of reduced immunogenicity when the antibodies are administered to humans.
  • a humanized monoclonal antibody comprises the variable domain of a murine antibody (or all or part of the antigen binding site thereof) and a constant domain derived from a human antibody.
  • a humanized antibody fragment may comprise the antigen binding site of a murine monoclonal antibody and a variable domain fragment (lacking the antigen-binding site) derived from a human antibody.
  • chimeric and further engineered monoclonal antibodies include those described in Riechmann et al., 1988, Nature 332:323, Liu et al., 1987, Proc. Nat. Acad. Sci. USA 84:3439, Larrick et al., 1989, Bio/Technology 7:934, and Winter et al., 1993, TIPS 14:139.
  • the chimeric antibody is a CDR grafted antibody.
  • mice in which one or more endogenous immunoglobulin genes have been inactivated by various means have been prepared.
  • Human immunoglobulin genes have been introduced into the mice to replace the inactivated mouse genes.
  • Antibodies produced in the animal incorporate human immunoglobulin polypeptide chains encoded by the human genetic material introduced into the animal. Examples of techniques for production and use of transgenic animals for the production of human or partially human antibodies are described in U.S. Patents 5,814,318, 5,569,825, and 5,545,806, which are incorporated by reference herein.
  • the present invention provides monoclonal antibodies that bind to a T R 2 cytokine or cytokine receptor.
  • Monoclonal antibodies may be produced using any technique known in the art, e.g., by immortalizing spleen cells harvested from the transgenic animal after completion of the immunization schedule.
  • the spleen cells can be immortalized using any technique known in the art, e.g., by fusing them with myeloma cells to produce hybridomas.
  • Myeloma cells for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency, and enzyme deficiencies that render them incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas).
  • Examples of suitable cell lines for use in mouse fusions include Sp-20, P3-X63/Ag8, P3-X63-Ag8.653, NSl/l.Ag 4 1, Sp210-Agl4, FO, NSO/U, MPC-11, MPCl 1-X45-GTG 1.7 and S194/5XX0 BuI; examples of cell lines used in rat fusions include R210.RCY3, Y3-Ag 1.2.3, IR983F and 4B210.
  • Other cell lines useful for cell fusions are U-266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6.
  • a hybridoma cell line is produced by immunizing an animal (e.g., a transgenic animal having human immunoglobulin sequences) with an immunogen; harvesting spleen cells from the immunized animal; fusing the harvested spleen cells to a myeloma cell line, thereby generating hybridoma cells; establishing hybridoma cell lines from the hybridoma cells, and identifying a hybridoma cell line that produces an antibody that binds to the desired target.
  • hybridoma cell lines, and antibodies produced therefrom are encompassed by the present invention.
  • Monoclonal antibodies secreted by a hybridoma cell line can be purified using any technique known in the art.
  • Hybridomas or mAbs may be further screened to identify mAbs with particular properties, such as the ability to block an IL-4- and/or an IL- 13 -induced activity.
  • Antibodies of the invention can comprise any constant region known in the art.
  • the light chain constant region can be, for example, a kappa- or lambda-type light chain constant region, e.g., a human kappa- or lambda-type light chain constant region.
  • the heavy chain constant region can be, for example, an alpha-, delta-, epsilon-, gamma-, or mu-type heavy chain constant regions, e.g., a human alpha-, delta-, epsilon-, gamma-, or mu-type heavy chain constant region.
  • the light or heavy chain constant region is a fragment, derivative, variant, or mutein of a naturally occurring constant region.
  • Antibodies of the invention include, but are not limited to, partially human and fully human monoclonal antibodies that inhibit a biological activity of IL-4 and also inhibit a biological activity of IL-13.
  • One embodiment is directed to a human monoclonal antibody that at least partially blocks binding of IL-4 to a cell, and at least partially blocks binding of IL-13 to a cell.
  • the antibodies are generated by immunizing a transgenic mouse with an IL-4 receptor immunogen.
  • the immunogen is a human IL-4 receptor polypeptide.
  • non-human antibodies of the invention can be, for example, derived from any antibody-producing animal, such as mouse, rat, rabbit, goat, donkey, or non-human primate (such as monkey (e.g., cynomologous or rhesus monkey) or ape (e.g., chimpanzee)).
  • Non-human antibodies of the invention can be used, for example, in in vitro and cell-culture based applications, or any other application where an immune response to the antibody of the invention does not occur or is insignificant, can be prevented, is not a concern, or is desired.
  • a non-human antibody of the invention is administered to a non-human subject.
  • the non-human antibody does not elicit an immune response in the non-human subject.
  • the non-human antibody is from the same species as the non- human subject, e.g., a mouse antibody of the invention is administered to a mouse.
  • An antibody from a particular species can be made by, for example, immunizing an animal of that species with the desired immunogen or using an artificial system for generating antibodies of that species (e.g., a bacterial or phage display-based system for generating antibodies of a particular species), or by converting an antibody from one species into an antibody from another species by replacing, e.g., the constant region of the antibody with a constant region from the other species, or by replacing one or more amino acid residues of the antibody so that it more closely resembles the sequence of an antibody from the other species.
  • the antibody is a chimeric antibody comprising amino acid sequences derived from antibodies from two or more different species.
  • the present invention provides antibodies that comprise a light chain variable region selected from the group consisting of L1-L6 and/or a heavy chain variable region selected from the group consisting of H1-H24, and fragments, derivatives, muteins, and variants thereof as described in US Pat. App. No. 07/0274996 Al, incorporated herein by reference in its entirety.
  • Such an antibody can be denoted using the nomenclature "LxHy”, wherein x corresponds to the number of the light chain variable region and y corresponds to the number of the heavy chain variable region.
  • L4H17 refers to an antibody with a light chain variable region comprising the amino acid sequence of L4 and a heavy chain variable region comprising the amino acid sequence of H 17.
  • Antibodies of the invention include, for example, L2H1, L3H1, L4H1, L5H1, L1H2, L1H3, L1H4, L1H5, L1H6, L1H7, L1H8, L1H9, LlHlO, LlHI l, L2H4, L2H12, L2H13, L2H14, L6H1, L2H2, L2H3, L2H6, L2H7, L2H8, L2H9, L2H10, and L2H11, as described in US Pat. App. No. 07/0274996 Al. Additional antibody variable sequences, e.g., human antibody variable sequences, also can be used.
  • the present invention provides an antibody comprising a light chain- variable domain comprising a sequence of amino acids that differs from the sequence of Ll only at one or more residues where any one of the sequences of L2-L6 differs from the sequence of Ll (e.g., said sequence of said antibody differs from the sequence of Ll at residue(s) 1, 4, 7 etc.).
  • said sequence of said light chain- variable domain comprises at least one amino acid residue of any of the sequence of any one of L2-L6 at a position where it differs from the sequence of Ll (e.g., said sequence comprises the residue(s) ElD, L4M, S7T, etc.).
  • said sequence differs from the sequence of Ll in at least one CDR (e.g., CDRl, CDR2, or CDR3).
  • said sequence differs from the sequence of Ll in at least one FR (e.g., FRl, FR2, FR3, or FR4).
  • the present invention provides an antibody comprising a heavy chain- variable domain comprising a sequence of amino acids that differs from the sequence of Hl only at one or more residues where any one of the sequences of H2- H24 differs from the sequence of Hl (e.g., said sequence of said antibody differs from the sequence of Hl at residue(s) 6, 13, 24 etc.).
  • said sequence of said heavy chain- variable domain comprises at least one amino acid residue of any of the sequence of any one of H2-H24 at a position where it differs from the sequence of Hl (e.g., said sequence comprises the residue(s) Q6E, H13Q, G24A, etc.).
  • said sequence differs from the sequence of Hl in at least one CDR (e.g., CDR2 or CDR3). In another embodiment, said sequence differs from the sequence of Hl in at least one FR (e.g., FRl or FR3).
  • the present invention provides methods of treating a gastrointestinal allergic condition in a subject.
  • Treatment encompasses, for example, alleviation or prevention of at least one symptom of a disorder, or reduction of disease severity, or the like.
  • a treatment need not effect a complete “cure”, or eradicate or improve every symptom or manifestation of a disease, to constitute a viable therapy.
  • drugs employed as therapeutic agents may reduce the severity of a given disease state, but need not abolish every manifestation of the disease to be regarded as useful therapeutic agents.
  • One embodiment of the invention is directed to a method comprising administering to a patient T H 2 cytokine antagonist in an amount and for a time sufficient to induce a sustained improvement over baseline of an indicator that reflects the severity of the particular disorder.
  • the method of treatment can comprise administering to the subject any type of treatment useful in achieving a particular therapeutic goal.
  • the treatment can comprise administering any kind of drug, compound, composition, pharmaceutical formulation, or the like. Examples of such substances include compounds that inhibit the activity a T R 2 cytokine or cytokine receptor. Illustrative examples of such compounds are provided herein.
  • drugs are administered to a subject in a manner appropriate to the indication. They may be administered by any suitable technique, including but not limited to parenterally, topically, orally, or by inhalation. If injected, the drug can be administered, for example, via intra-articular, intravenous, intramuscular, intralesional, interperitoneal or subcutaneous routes, by bolus injection, or continuous infusion.
  • Localized administration e.g. at a site of disease or injury is contemplated, as are transdermal delivery and sustained release from implants.
  • Delivery by inhalation includes, for example, nasal or oral inhalation, use of a nebulizer, inhalation of the antagonist in aerosol form, and the like.
  • Other alternatives include eyedrops; oral preparations including pills, syrups, lozenges or chewing gum; and topical preparations such as lotions, gels, sprays, and ointments.
  • drugs are administered in the form of a composition
  • a composition comprising at least one active substance and optionally one or more additional components such as a physiologically acceptable carrier, excipient or diluent.
  • Compositions may, for example, comprise a drug together with a buffer, antioxidant such as ascorbic acid, low molecular weight polypeptide (such as those having fewer than 10 amino acids), protein, amino acid, carbohydrate such as glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione, and other stabilizers and excipients.
  • a buffer such as ascorbic acid, low molecular weight polypeptide (such as those having fewer than 10 amino acids)
  • protein amino acid
  • carbohydrate such as glucose, sucrose or dextrins
  • chelating agents such as EDTA, glutathione, and other stabilizers and excipients.
  • Neutral buffered saline or saline mixed with conspecific serum albumin are examples of appropriate
  • preservatives such as benzyl alcohol may also be added.
  • the composition may be formulated as a lyophilizate using appropriate excipient solutions (e.g., sucrose) as diluents. Suitable components are nontoxic to recipients at the dosages and concentrations employed. Further examples of components that may be employed in pharmaceutical formulations are presented in Remington's
  • Kits for use by medical practitioners include a drug and a label or other instructions for use in treating any of the conditions discussed herein.
  • the kit preferably includes a sterile preparation of one or more drugs, which may be in the form of a composition as disclosed above, and may be in one or more vials.
  • Dosages and the frequency of administration may vary according to such factors as the route of administration, the particular drug employed, the nature and severity of the disease to be treated, whether the condition is acute or chronic, and the size and general condition of the subject. Appropriate dosages can be determined by procedures known in the pertinent art, e.g. in clinical trials that may involve dose escalation studies.
  • a drug may be administered, for example, once or more than once, e.g., at regular intervals over a period of time.
  • the drug is administered over a period of at least a month or more, e.g., for one, two, or three months or even indefinitely.
  • long-term treatment is generally most effective.
  • administration for shorter periods e.g. from one to six weeks, may be sufficient.
  • the drug is administered until the subjects manifests a medically relevant degree of improvement over baseline for the chosen indicator or indicators, or fails to show an expected or desired level of improvement over a particular period of time or number of treatments.
  • Examples of particular embodiments of the present invention involve administering a drug at a dosage of from about 1 ng of drug per kg of subject's weight per day ("lng/kg/day") to about 10 mg/kg/day, from about 500 ng/kg/day to about 5 mg/kg/day, and from about 5 ⁇ g/kg/day to about 2 mg/kg/day, to a subject.
  • a drug is administered to adults one time per week, two times per week, or three or more times per week, to treat a medical disorder disclosed herein. If injected, the effective amount of drug per adult dose may range from 1-20 mg/m 2 , and preferably is about 5-12 mg/ m 2 .
  • a flat dose may be administered; the amount may range from 5-100 mg/dose.
  • One range for a flat dose is about 20-30 mg per dose.
  • a flat dose of 25 mg/dose is repeatedly administered by injection. If a route of administration other than injection is used, the dose is appropriately adjusted in accordance with standard medical practices.
  • One example of a therapeutic regimen involves injecting a dose of about 20-30 mg of drug one to three times per week over a period of at least three weeks, though treatment for longer periods may be necessary to induce the desired degree of improvement.
  • one exemplary suitable regimen involves the subcutaneous injection of 0.4 mg/kg, up to a maximum dose of 25 mg of drug administered two or three times per week.
  • Particular embodiments of the methods provided herein involve subcutaneous injection of from 0.5 mg to 10 mg, preferably from 3 to 5 mg, of drug, once or twice per week.
  • Another embodiment is directed to pulmonary administration (e.g., by nebulizer) of 3 mg or more of drug once a week.
  • Examples of therapeutic regimens provided herein comprise subcutaneous injection of drug once a week, at a dose of 1.5 to 3 mg. Weekly administration of anti-IL-4R antibody is continued until a desired result is achieved, e.g., the subject's symptoms subside. Treatment may resume as needed, or, alternatively, maintenance doses may be administered.
  • a drug is administered to the subject in an amount and for a time sufficient to induce an improvement, preferably a sustained improvement, in at least one indicator that reflects the severity of the disorder that is being treated.
  • Various indicators that reflect the extent of the subject's illness, disease or condition may be assessed for determining whether the amount and time of the treatment is sufficient.
  • Such indicators include, for example, clinically recognized indicators of disease severity, symptoms, or manifestations of the disorder in question.
  • the degree of improvement generally is determined by a physician or other qualified caregiver, who may make this determination based on signs or symptoms, and who may also employ questionnaires that are administered to the subject, such as quality-of-life questionnaires developed for a given disease.
  • Elevated levels of T R 2 cytokines, or of IgE are associated with a number of disorders. Subjects may be screened, to identify those individuals who have elevated levels of such molecules, or to identify those with an elevated T H 2-type immune response, thereby identifying the subjects who may benefit most from treatment. Thus, treatment methods provided herein optionally comprise a first step of measuring a subject's level of a cytokine or IgE. Alternatively or additionally, a subject may be pre-screened to determine whether the subject has an elevated T ⁇ 2-type immune response, prior to administration of antibody(-ies) and/or antagonist(s) against one or more T ⁇ 2-type cytokines.
  • a subject's levels of aT ⁇ 2-type cytokine, or of IgE may be monitored during and/or after treatment, to detect changes in the levels of the cytokine or IgE.
  • the incidence of elevated cytokine or IgE, and the balance between T H 1 -type and T H 2-type immune responses may vary according to such factors as the stage of the disease or the particular form of the disease.
  • Known techniques may be employed for measuring these levels, e.g., in a subject's serum, and for assessing T H 2- type immune responses.
  • Cytokine levels in blood samples may be measured by ELISA, or by a LUMINEXTM multi-plex cytokine assay (Luminex Corporation,
  • anti-IL4R antibody and one or more additional IL-4R antagonists, for example, two or more antibodies or antibody derivatives of the invention, or an antibody or antibody derivative of the invention and one or more other IL-4R antagonists.
  • anti-IL-4R antibodies are administered alone or in combination with other agents useful for treating the condition with which the patient is afflicted. Examples of such agents include both proteinaceous and non- proteinaceous drugs. When multiple therapeutics are co-administered, dosages may be adjusted accordingly, as is recognized in the pertinent art.
  • “Co-administration" and combination therapy are not limited to simultaneous administration, but include treatment regimens in which an IL-4R antibody is administered at least once during a course of treatment that involves administering at least one other therapeutic agent to the patient.
  • agents that may be administered are other antibodies, cytokines, or cytokine receptors, which are chosen according to the particular condition to be treated.
  • non-proteinaceous drugs that are useful in treating one of the particular conditions discussed above may be administered.
  • Treatment may comprise administration of an IgE antagonist.
  • an IgE antagonist is an anti-IgE antibody, e.g., XOLAIR® (Genentech, South San Francisco, CA).
  • Humanized anti-IgE monoclonal antibodies are described in, for example, Presta et al, 1993, J. Immunol. 151 :2623-32 and Adelroth et al, 2000, J. Allergy Clin. Immunol. 106:253-59.
  • Treatment can also comprise administering an IL-5 antagonist, which may be a molecule that interferes with the binding of IL-5 to an IL-5 receptor, such as an anti- IL-5R or anti-IL-5 antibody (e.g., a human or humanized anti-IL-5 or anti-IL-5R monoclonal antibody), or a receptor such as a soluble human IL-5 receptor polypeptide.
  • an IL-5 antagonist which may be a molecule that interferes with the binding of IL-5 to an IL-5 receptor, such as an anti- IL-5R or anti-IL-5 antibody (e.g., a human or humanized anti-IL-5 or anti-IL-5R monoclonal antibody), or a receptor such as a soluble human IL-5 receptor polypeptide.
  • agents that can be used include anti-IL-4 antibodies, IL-4 muteins, IL-4 binding derivatives of IL-4R (as described in, e.g., U.S. Patents 5,840,869; 5,599,905, 5,856,296, 5,767,065, 5,717,072, 6,391,581, 6,548,655, 6,472,179, and 5,844,099), IL- 13 binding derivatives of IL-13, IL-4 and/or IL- 13 binding chimeric derivatives of IL-4R and IL- 13R, IL- 13 muteins, and antagonists of CD23 ⁇ e.g., anti-CD23 antibodies such as IDEC- 152TM (IDEC Pharmaceuticals, San Diego, CA), Phosphodiesterase 4 ⁇ e.g., ROFLUMILASTTM, Byk Gulden Pharmaceuticals, Konstanz, Germany), integrins ⁇ e.g., R411TM, Roche, Nutley, NJ), TIMs, Gob5,
  • Treatment also can comprise administering an IL-9 antagonist.
  • an IL-9 antagonist may be employed, such as an IL-9 receptor (e.g., a soluble form thereof), an antibody that interferes with binding of IL-9 to a cell surface receptor (e.g., an antibody that binds to IL-9 or to an IL-9 receptor polypeptide), or another compound that inhibits IL-9-induced biological activity.
  • IL-9 receptors include those described in WO 93/18047 and U.S. Patents 5,789,237 and 5,962,269, which are hereby incorporated by reference herein.
  • Treatment also can comprise administering any suitable IL- 13 antagonist, including but not limited to IL- 13 receptors (preferably soluble forms thereof), IL- 13 receptor antagonists, antibodies directed against IL- 13 or an IL- 13R, other proteins that interfere with the binding of IL- 13 to an IL- 13R, and compounds that inhibit IL- 13 -mediated signal transduction.
  • IL- 13 receptors and heterodimers comprising IL- 13R polypeptides as components thereof are described above.
  • Antibodies that are raised against IL-4R may be screened for the ability to also function as IL- 13 antagonists.
  • Another method provided herein comprises co-administering an antibody or antibody derivative of the invention and interferon- ⁇ (IFN- ⁇ ).
  • IFN- ⁇ interferon- ⁇
  • Treatment also can comprise administering one or more leukotriene receptor antagonists.
  • leukotriene receptor antagonists include but are not limited to montelukast (e.g., SINGULAIR®, Merck & Co., Whitehouse, NJ), pranlukast (e.g., ONONTM Ono Pharmaceuticals, Osaka, Japan), and zafirlukast (e.g., ACCOLATETM, AstraZeneca, Wilmington, DE).
  • Drugs that function as 5 -lipoxygenase inhibitors may be co-administered with an IL-4R antagonist to treat asthma.
  • Treatment may comprise administering an IL- 17 antagonist, including but not limited to an IL- 17 receptor (e.g., soluble forms thereof), IL- 17 receptor antagonists, antibodies directed against IL- 17 or an IL- 17 receptor, other proteins that interfere with the binding of IL- 17 to an IL- 17 receptor, and compounds that inhibit IL- 17- mediated signal transduction.
  • an IL- 17 antagonist including but not limited to an IL- 17 receptor (e.g., soluble forms thereof), IL- 17 receptor antagonists, antibodies directed against IL- 17 or an IL- 17 receptor, other proteins that interfere with the binding of IL- 17 to an IL- 17 receptor, and compounds that inhibit IL- 17- mediated signal transduction.
  • An IL- 17 receptor including soluble forms thereof and oligomers thereof, is described in WO 96/29408, hereby incorporated by reference.
  • Treatment may also comprise administering a TNF antagonist, including but not limited to a TNF receptor (preferably soluble forms thereof), fusion proteins comprising a TNF receptor (or comprising the TNF-binding portion of a TNF receptor), TNF receptor antagonists, antibodies directed against TNF or a TNF receptor, other proteins that interfere with the binding of TNF to a TNF receptor, and compounds that inhibit TNF -mediated signal transduction.
  • a TNF antagonist including but not limited to a TNF receptor (preferably soluble forms thereof), fusion proteins comprising a TNF receptor (or comprising the TNF-binding portion of a TNF receptor), TNF receptor antagonists, antibodies directed against TNF or a TNF receptor, other proteins that interfere with the binding of TNF to a TNF receptor, and compounds that inhibit TNF -mediated signal transduction.
  • TNF inhibitors are the drugs thalidomide and pentoxyfylline.
  • the TNF receptor protein known as p75 or p80 TNF-R preferably is employed.
  • a preferred TNF antagonist is a soluble human TNF receptor (sTNF-R) in dimeric form, such as dimers of sTNF-R/Fc fusion proteins.
  • sTNF-R soluble human TNF receptor
  • dimer is etanercept (ENBREL®, Immunex Corporation, Seattle, WA).
  • ENBREL® etanercept
  • p75/p80 TNF-R including soluble fragments and other forms thereof, is described in WO 91/03553, hereby incorporated by reference herein.
  • cytokine antagonists and other agents/drugs are disclosed herein as being useful for combination therapy. It is to be understood that such antagonists, agents, or drugs also find use as single agents in the methods of treatment of the invention. It also is to be understood that disclosure of methods involving administration of an antagonist to a particular cytokine, to treat a disease, encompasses administration of one type of antagonist, and also encompasses administration of two or more different antagonists for that cytokine, unless specified otherwise.
  • the method of treatment can further comprise the administration of other agents.
  • These other agents can be administered for any medically indicated purpose, for example, to further reduce a T ⁇ 2-associated activity, or to provide symptomatic relief or palliative care, or to manage a side-effect or other undesirable effect of treatment.
  • mice were housed under pathogen- free conditions and treated in a humane manner according to institutional guidelines. Water was available continuously through automatic ports and a commercial mouse diet was provided ad libitum.
  • mice were given 50 ⁇ g ovalbumin (OVA grade V; A- 5503; Sigma-Aldrich, St.Louis, MO) interperitoneally (i.p.) in the presence of 1 mg of an aluminum potassium sulfate adjuvant (alum: ALK(SO 4 ⁇ - 12H 2 O; A-7210; Sigma- Aldrich, Milwaukee, WI).
  • OVA grade V ovalbumin
  • A-7210 aluminum potassium sulfate adjuvant
  • the mice were sensitized twice i.p., two weeks apart, with the OV A/adjuvant.
  • mice were deprived of food for three to four hours prior to each intragastric challenge. Three times a week, mice were held in the supine position and orally administered 10 or 50 mg of soluble OVA dissolved in 250 ⁇ l of 0.9% sterile saline. Challenges were performed with intragastric feeding needles (22G - 1.5 in - 1.25 mm ball; 01-290-2B; Fisher Scientific Co, Pittsburg, PA).
  • mice Two milligrams of a rat anti-mouse IL-4R ⁇ antibody or of a sham rat IgGl antibody were administered i.p. to the mice either twenty-four hours prior to the first intragastric OVA challenge, or after they had developed diarrhea.
  • Diarrhea was assessed visually by closely monitoring mice for 1 hour following oral allergen challenge.
  • Ribonuc lease protection assay Jejunal RNA was obtained using Trizol reagent (Gibco-BRL, Grand-Island, NY) following the manufacturer's protocol. The RPA was performed by making a radioactive probe from the mCk-lb multiprobe templet (Riboquant multi-probe RPA system, BD Biosciences-Pharmingen, San Diego, CA). RNA from OVA and saline challenged mice was then hybridized overnight with the radioactive probe, purified and finally run on an Urea-Acrylamide gel at 75 W as described in the Riboquant protocol from Pharmingen.
  • In vivo cytokine capture assay The in vivo cytokine capture assay (IVCCA) was used to monitor in vivo production of IL-4. Briefly, mice were injected intravenously with 200 ⁇ l of biotinylated anti-mouse IL-4 (BVD4-1D11; lO ⁇ g) monoclonal antibody. Mice were bled 24 h later and plasma levels of captured IL-4 were measured by ELISA using plates coated with non-neutralizing monoclonal antibodies directed either against IL4 (BVD6-24G2.3). Complexes formed by recombinant cytokines and anti-cytokine monoclonal antibodies were used as standards to calculate plasma cytokine concentrations.
  • IVCCA in vivo cytokine capture assay
  • Intestinal mast cell quantification Mice were sacrificed 60-90 min after the last OVA exposure. Upper jejunum tissue was collected, fixed in 10% formalin, processed by using standard histological techniques. The 5 ⁇ m tissue sections were also stained for mucosal mast cells with chloroacetate esterase and lightly conterstained with methyl green. At least three random sections per mouse were analyzed. Quantification of stained cells per square millimeter was performed by morphometric analysis using the Metamorph Imaging System (Universal Imaging Corporation, West Chester, PA).
  • mice Data are expressed as mean ⁇ SEM. Statistical significance comparing different sets of mice was determined by Student's unpaired t- test or the non-parametric Mann Whitney U-test.
  • mice treated with antibody before the first intragastric challenge after five exposures, all control mice had developed diarrhea compared to only one out of five mice given the anti-IL-4R ⁇ antibody. This delay in the development of allergic diarrhea was associated with lower plasma IgE levels as well as impaired intestinal mast cell accumulation and degranulation as suggested by decreased mMCP-1 plasma levels.
  • mice treated with antibody after they developed diarrhea showed significantly reduced total IgE levels, mMCP-1 levels, and mast cells, relative to control mice, within a week. Allergic diarrhea development did not appear to be affected.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des procédés et des compositions pour traiter des troubles allergiques gastro-intestinaux. Ces traitements comprennent, par exemple, des procédés prophylactiques. Des exemples de procédés prophylactiques comprennent des procédés qui préviennent la survenue, ou qui réduisent le risque de survenue, ou qui réduisent la durée ou l'intensité, d’un ou de plusieurs symptômes ou effets de la maladie allergique gastro-intestinale. Les procédés de l’invention comprennent également, par exemple, des procédés pour traiter un sujet qui présente un ou plusieurs symptômes ou effets d’une maladie allergique gastro-intestinale.
PCT/US2009/037192 2008-03-13 2009-03-13 Compositions et procédés relatifs à des états d’allergie gastro-intestinale WO2009114828A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3641208P 2008-03-13 2008-03-13
US61/036,412 2008-03-13

Publications (1)

Publication Number Publication Date
WO2009114828A1 true WO2009114828A1 (fr) 2009-09-17

Family

ID=40847576

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/037192 WO2009114828A1 (fr) 2008-03-13 2009-03-13 Compositions et procédés relatifs à des états d’allergie gastro-intestinale

Country Status (1)

Country Link
WO (1) WO2009114828A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020135471A1 (fr) 2018-12-25 2020-07-02 Qyuns Therapeutics Co., Ltd. Anticorps monoclonal contre le récepteur alpha de l'interleukine-4 humaine et son utilisation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040037909A1 (en) * 2002-08-23 2004-02-26 Kim Bong Cheol Composition comprising the extract of actinidia arguta and related species for the prevention and treatment of allergic disease and non-allergic inflammatory disease
EP1716851A1 (fr) * 2004-02-17 2006-11-02 Ishihara Sangyo Kaisha, Ltd. Thioamides et sels de ceux-ci et inhibiteurs de production de cytokines contenant les deux
US20070048328A1 (en) * 2005-08-31 2007-03-01 Yamanashi University Allergy prevention method or treatment method, diet and oral drug

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040037909A1 (en) * 2002-08-23 2004-02-26 Kim Bong Cheol Composition comprising the extract of actinidia arguta and related species for the prevention and treatment of allergic disease and non-allergic inflammatory disease
EP1716851A1 (fr) * 2004-02-17 2006-11-02 Ishihara Sangyo Kaisha, Ltd. Thioamides et sels de ceux-ci et inhibiteurs de production de cytokines contenant les deux
US20070048328A1 (en) * 2005-08-31 2007-03-01 Yamanashi University Allergy prevention method or treatment method, diet and oral drug

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ADEL-PATIENT K ET AL: "Evaluation of a high IgE-responder mouse model of allergy to bovine beta-lactoglobulin (BLG): development of sandwich immunoassays for total and allergen-specific IgE, IgG1 and IgG2a in BLG-sensitized mice", JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, vol. 235, no. 1-2, 1 February 2000 (2000-02-01), pages 21 - 32, XP004188228, ISSN: 0022-1759 *
ITO KAZUE ET AL: "Murine model of IgE production with a predominant Th2-response by feeding protein antigen without adjuvants", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 27, no. 12, December 1997 (1997-12-01), pages 3427 - 3437, XP002537268, ISSN: 0014-2980 *
RUPA P ET AL: "Engineered recombinant ovomucoid third domain can modulate allergenic response in Balb/c mice model", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 342, no. 3, 14 April 2006 (2006-04-14), pages 710 - 717, XP024923640, ISSN: 0006-291X, [retrieved on 20060414] *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020135471A1 (fr) 2018-12-25 2020-07-02 Qyuns Therapeutics Co., Ltd. Anticorps monoclonal contre le récepteur alpha de l'interleukine-4 humaine et son utilisation

Similar Documents

Publication Publication Date Title
US20210079105A1 (en) Methods of Treating Inflammatory Conditions
KR101225463B1 (ko) 인터류킨-4 수용체에 결합하는 항체
ES2549767T3 (es) Uso de anticuerpos de interleuquina-4 y composiciones de los mismos
JP5255453B2 (ja) Par−2に結合する抗体
US9718884B2 (en) Anti-TNF-/anti-IL-23 bispecific antibodies
CA2685123A1 (fr) Procedes et compositions pour traiter et surveiller le traitement des troubles associes a il-13
CA2672215A1 (fr) Methodes et compositions pour le traitement et le controle d'un traitement de troubles associes a il-13
KR20140061403A (ko) 항―il―13 항체를 이용하여 천식을 치료하기 위한 방법 및 조성물
DK2340039T3 (en) Use of IL-20 antagonists for treating osteoporosis
CN112424226A (zh) 用于治疗重度哮喘的抗cd6抗体
KR102427097B1 (ko) 항-tnf-알파 항체 요법을 받은 건선 환자의 치료 방법
TW202304980A (zh) 經修飾的抗tslp抗體
Guilleminault et al. Pharmacological approaches to target type 2 cytokines in asthma
JP2012131825A (ja) Th2介在性状態の治療のためのKIM−1抗体
TW202332691A (zh) Ige調節的疾病的治療
WO2009114828A1 (fr) Compositions et procédés relatifs à des états d’allergie gastro-intestinale
US20220267432A1 (en) Method for treating autoimmune disease by il-17 antagonist
EA043630B1 (ru) Способы лечения воспалительных состояний
NZ619873B2 (en) Methods and compositions for treating asthma using anti-il-13 antibodies

Legal Events

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

Ref document number: 09719063

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09719063

Country of ref document: EP

Kind code of ref document: A1