US20170145089A1 - Anti-il4-il 13 bispecific antibodies - Google Patents

Anti-il4-il 13 bispecific antibodies Download PDF

Info

Publication number
US20170145089A1
US20170145089A1 US15/320,106 US201515320106A US2017145089A1 US 20170145089 A1 US20170145089 A1 US 20170145089A1 US 201515320106 A US201515320106 A US 201515320106A US 2017145089 A1 US2017145089 A1 US 2017145089A1
Authority
US
United States
Prior art keywords
antibody
dose
protein
fragment
tarc
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
US15/320,106
Other languages
English (en)
Inventor
Corrine ESPERET
Alexandre JAGERSCHMIET
Christina SOUBRANE
Arun Subramaniam
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.)
Sanofi SA
Original Assignee
Sanofi SA
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 Sanofi SA filed Critical Sanofi SA
Priority to US15/320,106 priority Critical patent/US20170145089A1/en
Publication of US20170145089A1 publication Critical patent/US20170145089A1/en
Assigned to SANOFI reassignment SANOFI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ESPERET, CORINNE, JAGERSCHMIDT, ALEXANDRE, SOUBRANE, Christina, SUBRAMANIAM, ARUN
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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/247IL-4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • 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
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/521Chemokines
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • Interleukin-4 is a pleiotropic cytokine that has a broad spectrum of biological effects on lymphoid B and T cells, and many non-lymphoid cells including monocytes, endothelial cells and fibroblasts.
  • IL-4 induces the expression of class II major histocompatibility complex molecules on resting B cells, and enhances the secretion of IgG4 and IgE by human B cells.
  • IL-4 is associated with a Th2-type immune response, and is produced by and promotes differentiation of Th2 cells.
  • IL-4 has been implicated in a number of disorders, such as allergy and asthma.
  • IL-13 is a cytokine of 112 amino acids secreted by the activated T lymphocytes, the B lymphocytes and the mastocytes after activation (Minty, A. et al., Nature, 1993, 362, 248-250, and McKenzie, A. N. et al., Proc. Natl. Acad. Sci. U.S.A, 1993, 90, 3735-3739).
  • IL-13 has been described as an IL-4-like cytokine. Its activities are indeed similar to those of IL-4 on the B cells (Defrance, T. et al., J. Exp. Med., 1994, 179, 135-143, Punnonen, J.
  • IL-13 various biological activities of IL-13 on the monocytes/macrophages, the B lymphocytes and certain haematopoietic precursors have been described in detail by A. J. Minty as well as in review articles on IL-13.
  • Several data indicate, in addition, that this cytokine has a pleiotropic effect on other cell types.
  • These non-haematopoietic cells which are directly affected by IL-13 are endothelial and microglial cells, keratinocytes and kidney and colon carcinomas.
  • IL-13 and IL-4 have a common receptor, or at the very least some of the components of a common receptor complex, as well as common signal transduction elements (Zurawski S. M. et al., EMBO J., 1993, 12, 2663-2670, Aversa, G. et al., J. Exp. Med., 1993, 178, 2213-2218, Vita, N. et al., J. Biol. Chem., 1995, 270, 3512-3517, Lefort, S.
  • This receptor is present at the surface of various cell types, in a variable number according to the cell type considered.
  • the comparative distribution of the IL-13 and IL-4 receptors has been indicated by A. J. Minty (Interleukin-13 for Cytokines in Health and Disease. Eds D. G. Remick and J. S. Frie, Marcel Decker, N.Y. 1996).
  • the cell surface receptors and receptor complexes bind IL-4 and/or IL-13 with different affinities.
  • the principle components of receptors and receptor complexes that bind IL-4 and/or IL-13 are IL-4R ⁇ , IL-13R ⁇ l and IL-13R ⁇ 2. These chains are expressed on the surface of cells as monomers or heterodimers of IL-4R ⁇ /IL-13R ⁇ l (Type II IL-4R) or IL-4R ⁇ /c (Type I IL-4R).
  • IL-4R ⁇ monomer and IL-4R ⁇ /c heterodimer bind IL-4, but not IL-13.
  • IL-13R ⁇ 1 and IL-13R ⁇ 2 monomers bind IL-13, but do not bind IL-4.
  • IL-4R ⁇ /IL-13R ⁇ 1 heterodimer binds both IL-4 and IL-13 (Murata et al., Int. J. Hematol., 1999, 69, 13-20).
  • Th2-type immune responses promote antibody production and humoral immunity, and are elaborated to fight off extracellular pathogens.
  • Th2 cells are mediators of Ig production (humoral immunity) and produce IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13 (Tanaka, et al., Cytokine Regulation of Humoral Immunity, 251-272, Snapper, ed., John Wiley and Sons, New York (1996)).
  • Th2-type immune responses are characterized by the generation of certain cytokines (e.g., IL-4, IL-13) and specific types of antibodies (IgE, IgG4) and are typical of allergic reactions, which may result in watery eyes and asthmatic symptoms, such as airway inflammation and contraction of airway muscle cells in the lungs.
  • cytokines e.g., IL-4, IL-13
  • IgE specific types of antibodies
  • IL-4 and IL-13 are therapeutically important cytokines based on their biological functions and play critical roles in many diseases.
  • IL-4 has been shown to be able to inhibit autoimmune disease and IL-4 and IL-13 have both shown the potential to enhance anti-tumor immune responses.
  • Elevations in IL-4 and IL-13 and their receptors have been linked to the pathogenesis of idiopathic pulmonary fibrosis (IPF) (Jakubzick et al., Am J Pathol. (2004) 164:1989-2001; Murray et al. Int J Biochem Cell Biol. (2008) 40:2174-82).
  • IPF idiopathic pulmonary fibrosis
  • TH2 cytokines IL-4 and IL-13 play multiple roles in the pathogenesis of IPF as mediators of this lung tissue remodeling and fibrosis.
  • Th2-type CD4+ T cells in the lung are likely the predominant sources of IL-4 and IL-13, and are implicated as important regulators of extracellular matrix remodeling (Wynn, Nat. Rev. Immunol, (2004) 4:583-594), other cell types including mast cells, basophils, eosinophils, macrophages and epithelial cells may also be potential sources of these cytokines (Gordon and Martinez, Immunity Rev. (2010) 32: 593-604).
  • IL-13 and IL-4 levels in bronchial alveolar lavage fluid are elevated compared to normal controls.
  • therapies capable of suppressing or neutralizing these cytokines have the potential for delaying the progression of fibrosis in IPF patients. Since both cytokines are involved in the pathogenesis of allergic diseases or fibrotic diseases, inhibitors of these cytokines could provide therapeutic benefits.
  • the invention provides safe doses in a human subject of a dual-V-region antibody-like protein or a fragment thereof that specifically binds to IL-4 and IL-13, wherein the dose comprises up to about 200 mg of the antibody-like protein or fragment thereof.
  • the human subject has idiopathic pulmonary fibrosis (IPF).
  • IPF idiopathic pulmonary fibrosis
  • the safe dose comprises about 200 mg of the antibody-like protein or fragment thereof.
  • the safe dose comprises about 100 mg of the antibody-like protein or fragment thereof.
  • the safe dose comprises about 50 mg of the antibody-like protein or fragment thereof.
  • the safe dose is administered once weekly. In some embodiments, the safe dose is administered subcutaneously.
  • the invention provides methods of determining whether a dose comprising a dual-V-region antibody-like protein or a fragment thereof administered to a human subject specifically binds to IL-4 or IL-13 within the human subject, the method comprising: (a) administering the dose to the human subject; and (b) measuring the amount of TARC/CCL17 protein in a blood, plasma, or serum sample drawn from the human subject, wherein a decrease in the amount of TARC/CCL17 in the blood sample relative to an amount of TARC/CCL17 in the subject measured before the dose was administered signifies binding of the dual-V-region antibody-like protein or fragment thereof to IL-4 or IL-13.
  • the methods further comprise step (c): increasing or decreasing the dose depending on the magnitude of the decrease in TARC/CCL17 measured in step (b).
  • step (c) further comprises increasing the dose if the decrease in TARC/CCL17 measured in step (b) is below a threshold value, or decreasing the dose if the decrease in TARC/CCL17 measured in step (b) is above a threshold value.
  • the threshold value of step (c) is about a 20% to about a 60% decrease in the amount of TARC/CCL17 relative to the amount of TARC/CCL17 in the subject measured before the dose was administered.
  • the threshold value is about a 40% to about a 50% decrease in the amount of TARC/CCL17 relative to the amount of TARC/CCL17 in the subject measured before the dose was administered.
  • the threshold value is about a 43% decrease in the amount of TARC/CCL17 relative to the amount of TARC/CCL17 in the subject measured before the dose was administered.
  • the dose is administered subcutaneously.
  • the amount of TARC/CCL17 is detected in step (b) by enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • the human subject has idiopathic pulmonary fibrosis (IPF).
  • the invention provides a protein biomarker for binding of an antibody or antibody-like binding protein or fragment thereof to IL-4 or IL-13 or both in a human subject, wherein the biomarker is TARC/CCL17.
  • the antibody or antibody-like binding protein or fragment thereof is a dual-V-region antibody-like binding protein or fragment thereof.
  • the human subject has idiopathic pulmonary fibrosis (IPF).
  • the invention provides methods of treating idiopathic pulmonary fibrosis (IPF), comprising administering to a human subject with IPF up to 200 mg of a dual-V-region antibody-like binding protein or a fragment thereof.
  • IPF idiopathic pulmonary fibrosis
  • the dual-V-region antibody-like binding protein or fragment thereof binds IL-4, IL-13, or both IL-4 and IL-13.
  • the dual-V-region antibody-like binding protein or fragment thereof is administered once per week.
  • the dual-V-region antibody-like binding protein or fragment thereof is administered subcutaneously.
  • the invention provides uses of a safe dose of a dual-V-region antibody-like binding protein or a fragment thereof for the treatment of idiopathic pulmonary fibrosis (IPF).
  • the safe dose is up to 200 mg of the dual-V-region antibody-like binding protein or a fragment thereof.
  • the safe dose is about 50 mg, or about 100 mg, or about 200 mg of the dual-V-region antibody-like binding protein or a fragment thereof.
  • the safe dose is administered subcutaneously.
  • the safe dose is administered once per week.
  • the dual-V-region antibody-like binding protein or a fragment thereof binds IL-4, IL-13, or both IL-4 and IL-13.
  • the invention provides safe doses of a dual-V-region antibody-like binding protein or a fragment thereof for the treatment of idiopathic pulmonary fibrosis (IPF).
  • the safe dose is up to 200 mg of the dual-V-region antibody-like binding protein or a fragment thereof.
  • the safe dose is about 50 mg, or about 100 mg, or about 200 mg of the dual-V-region antibody-like binding protein or a fragment thereof.
  • the safe dose is administered subcutaneously.
  • the safe dose is administered once per week.
  • the dual-V-region antibody-like binding protein or a fragment thereof binds IL-4, IL-13, or both IL-4 and IL-13.
  • FIG. 1 shows the relationship of IL-4 and IL-13 to TARC/CCL17 signaling.
  • FIG. 2 shows a trend of reduced TARC/CCL17 expression in the blood of human subjects with increasing doses of SAR156597.
  • FIG. 3 shows persistent reduction in TARC/CCL17 expression in the blood of human subjects 18 weeks following the first administration of SAR156597.
  • the graph shows the average amount of TARC/CCL17 versus time in the blood of patients administered 50 mg, 100 mg, or 200 mg SAR156597 SC once weekly.
  • FIGS. 5A-5C shows fibrotic changes in the lungs of FRA-2 overexpressing transgenic mice over time.
  • FIG. 6 shows images of lung samples of FRA-2 overexpressing mice at 9, 14, and 17 weeks.
  • FIG. 7 shows collagen content, as measured by hydroxyproline) in the skin of FRA2 mice at 13 and 16 weeks compared to wild-type control.
  • FIG. 8 shows the increased dermal thickness in samples from FRA2 mice at 8, 13, and 16 weeks compared to wild-type control.
  • FIG. 9 shows a cytokine profile analysis of the developing and fibrosing lungs and skin.
  • FIG. 10 shows a gene expression analysis of the lungs and skin from FRA2 mice, showing an increase in key signatory fibrotic markers.
  • FIG. 11 shows hydroxyproline levels in IL-13 Ab treated FRA2 mouse lungs.
  • FIG. 12 shows histopathology analysis of FRA2 and littermate control lungs, the results of which are quantitated in FIG. 13 .
  • FIG. 14 shows transcript analysis by real-time PCR for IPF biomarkers FN1, SPDEF, and MUC5B.
  • FIG. 15 shows transcript analysis by real-time PCR for profibrotic markers CCL2, CCL11, and CCL22.
  • FIG. 16 shows transcript analysis by real-time PCR for IL-6, ARG1, and IL13Ra2.
  • FIG. 17 shows IL-13, IL-4, and IL-17 protein expression levels in lung homogenates, as analyzed by ELISA.
  • FIG. 18 shows MCP-1 (CCL2), CCL17, and YKL-40 protein expression levels in lung homogenates, as analyzed by ELISA.
  • FIG. 19A shows the transgenic constructs for ectopic expression of Fra-2 in vivo as used in Eferl et al., 2008, Proc. Natl. Acad. Sci. 105(30): 10525-10530; and FIG. 19B shows the Fra-2 transgenic vector used herein, as described in Example 5.
  • the term “substantially” is utilized herein to represent the inherent degree of uncertainty that can be attributed to any quantitative comparison, value, measurement, or other representation.
  • the term “substantially” is also utilized herein to represent the degree by which a quantitative representation can vary from a stated reference without resulting in a change in the basic function of the subject matter at issue.
  • polypeptide As used herein, the terms “polypeptide,” “protein,” and “peptide” are interchangeable and refer to a chain of amino acid monomers linked by peptide bonds. Typically, polypeptide chains are unbranched. As used herein, the terms “residue” and “protein residue” are interchangeable and refer to an amino acid that is bonded with other amino acids by one or more peptide bonds within a protein.
  • Interleukin-4 (IL-4) relates to the naturally occurring, or endogenous mammalian IL-4 proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian IL-4 protein ⁇ e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature IL-4 protein, polymorphic or allelic variants, and other isoforms of an IL-4 and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated).
  • Naturally occurring or endogenous IL-4 includes wild type proteins such as mature IL-4, polymorphic or allelic variants and other isoforms and mutant forms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces IL-4, for example. These proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding IL-4, arc referred to by the name of the corresponding mammal.
  • the protein is designated as a human IL-4.
  • IL-4 Several mutant IL-4 proteins are known in the art, such as those disclosed in WO 03/038041.
  • Interleukin-13 refers to naturally occurring or endogenous mammalian IL-13 proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian IL-13 protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature IL-13 protein, polymorphic or allelic variants, and other isoforms of IL-13 (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., Hpidated, glycosylated).
  • Naturally occurring or endogenous IL-13 include wild type proteins such as mature IL-13, polymorphic or allelic variants and other isoforms and mutant forms which occur naturally in mammals (e.g., humans, non-human primates).
  • IL-13 encompasses the human IL-13 variant in which Arg at position 110 of mature human IL-13 is replaced with Gin (position 110 of mature IL-13 corresponds to position 130 of the precursor protein) which is associated with asthma (atopic and nonatopic asthma) and other variants of IL-13.
  • Such proteins can be recovered or isolated from a source which naturally produces IL-13, for example.
  • proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding IL-13 are referred to by the name of the corresponding mammal.
  • the protein is designated as a human IL-13.
  • mutant IL-13 proteins are known in the art, such as those disclosed in WO 03/035847.
  • the invention relates to the treatment of idiopathic pulmonary fibrosis (IPF).
  • IPF idiopathic pulmonary fibrosis
  • IL-4 and IL-13 are therapeutically important cytokines based on their biological functions and play critical roles in many diseases, including asthma (Curr Opin Allergy Clin Immunol 2005, Vo. 5, 161-166).
  • IL-4 has been shown to be able to inhibit autoimmune disease and IL-4 and IL-13 have both shown the potential to enhance anti-tumor immune responses. Elevations in IL-4 and IL-13 and their receptors have been linked to the pathogenesis of idiopathic pulmonary fibrosis (IPF) (Jakubzick C. et al., Am J Pathol.
  • TH2 cytokines IL-4 and IL-13 play multiple roles in the pathogenesis of IPF as mediators of this lung tissue remodeling and fibrosis (Wynn, T A, Naat. Rev. Immunol, 4:583-594, 2004) and other cell types including mast cells, basophils, eosinophils, macrophages and epithelial cells may also be potential sources of these cytokines (Gordon S and Martinez F O, Immunity Rev. 32:593-604, 2010).
  • IL-13 and IL-4 levels in bronchial alveolar lavage fluid are elevated compared to normal controls.
  • therapies capable of suppressing or neutralizing these cytokines have the potential for delaying the progression of fibrosis in IPF patients. Since both cytokines are involved in the pathogenesis of allergic diseases or fibrotic diseases, inhibitors of these cytokines could provide therapeutic benefits.
  • substantially identical with respect to an antibody chain polypeptide sequence may be construed as an antibody chain exhibiting at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or more sequence identity to the reference polypeptide sequence.
  • the term with respect to a nucleic acid sequence may be construed as a sequence of nucleotides exhibiting at least about 85%, 90%, 95%, 96%, 97%, 98%, 99% or more sequence identity to the reference nucleic acid sequence. Identity can be determined by using any bioinformatics tool available to one skilled in the art. For example, Basic Local Alignment Search Tool (BLAST) is commonly employed to determine sequence identity (Altschul et al., Journa J. Mol. Biol. (1990) 215:403-410).
  • BLAST Basic Local Alignment Search Tool
  • identity or “homology” may mean the percentage of nucleotide bases or amino acid residues in the candidate sequence that are identical with the residue of a corresponding sequence to which it is compared, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity for the entire sequence, and not considering any conservative substitutions as part of the sequence identity. Neither N-terminal or C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are available and well known in the art. Sequence identity may be measured using sequence analysis software.
  • “Substitutional” variants are those that have at least one amino acid residue in a native sequence removed and replaced with a different amino acid inserted in its place at the same position.
  • the substitutions may be single, where only one amino acid in the molecule is substituted, or may be multiple, where two or more amino acids are substituted in the same molecule.
  • the plural substitutions may be at consecutive sites.
  • one amino acid can be replaced with plural residues, in which case such a variant comprises both a substitution and an insertion.
  • “Insertional” variants are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a native sequence. Immediately adjacent to an amino acid means connected to either the ⁇ -carboxyl or ⁇ -amino functional group of the amino acid.
  • “Deletional” variants are those with one or more amino acids in the native amino acid sequence removed. Ordinarily, deletional variants will have one or two amino acids deleted in a particular region of the molecule.
  • antibody is used in the broadest sense, and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), antibody fragments or synthetic polypeptides carrying one or more CDR or CDR-derived sequences so long as the polypeptides exhibit the desired biological activity.
  • Antibodies (Abs) and immunoglobulins (Igs) are glycoproteins having the same structural characteristics. Generally, antibodies are considered Igs with a defined or recognized specificity. Thus, while antibodies exhibit binding specificity to a specific target, immunoglobulins include both antibodies and other antibody-like molecules which lack target specificity.
  • the antibodies of the invention can be of any class (e.g., IgG, IgE, IgM, IgD, IgA and so on), or subclass (e.g., IgG 1 , IgG 2 , IgG 2a , IgG 3 , IgG 4 , IgA 1 , IgA 2 and so on) (“type” and “class”, and “subtype” and “subclass”, are used interchangeably herein).
  • Native or wildtype that is, obtained from a non-artificially manipulated member of a population, antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains.
  • Each heavy chain has at one end a variable domain (V H ) followed by a number of constant domains.
  • Each light chain has a variable domain at one end (V L ) and a constant domain at the other end.
  • non-artificially manipulated is meant not treated to contain or express a foreign antigen binding molecule.
  • Wildtype can refer to the most prevalent allele or species found in a population or to the antibody obtained from a non-manipulated animal, as compared to an allele or polymorphism, or a variant or derivative obtained by a form of manipulation, such as mutagenesis, use of recombinant methods and so on to change an amino acid of the antigen-binding molecule.
  • anti-IL-4 antibody means an antibody or polypeptide derived therefrom (a derivative) which binds specifically to IL-4 as defined herein, including, but not limited to, molecules which inhibit or substantially reduce the binding of IL-4 to its receptor or inhibit IL-4 activity.
  • anti-IL-13 antibody means an antibody or polypeptide derived therefrom (a derivative) which binds specifically to IL-13 as defined herein, including, but not limited to, molecules which inhibit or substantially reduce the binding of IL-13 to its receptor or inhibit IL-13 activity.
  • variable in the context of a variable domain of antibodies refers to certain portions of the pertinent molecule which differ extensively in sequence between and among antibodies and are used in the specific recognition and binding of a particular antibody for its particular target. However, the variability is not evenly distributed through the variable domains of antibodies. The variability is concentrated in three segments called complementarity determining regions (CDRs; i.e., CDR1, CDR2, and CDR3) also known as hypervariable regions, both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework (FR) regions or sequences.
  • CDRs complementarity determining regions
  • variable domains of native heavy and light chains each comprise four FR regions, largely adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together often in proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the target (epitope or determinant) binding site of antibodies (see Kabat et al. Sequences of Proteins of Immunological Interest, National Institute of Health, Bethesda, Md. (1987)).
  • numbering of immunoglobulin amino acid residues is done according to the immunoglobulin amino acid residue numbering system of Kabat et al., unless otherwise indicated.
  • One CDR can carry the ability to bind specifically to the cognate epitope.
  • hinge or “hinge region” as used in the present invention refers to the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.
  • an antibody, antigen, or antigen-binding protein is a compound or molecule having qualitative biological activity in common with a full-length antibody or antigen of interest.
  • a functional fragment or analog of an anti-IL-4 antibody is one that can bind to an IL-4 molecule, or that can prevent or substantially reduce the ability of a ligand, or an agonistic or antagonistic antibody, to bind to IL-4.
  • fragment and “antibody fragment” refer to a portion of an intact or a full-length chain or an antibody, generally the target binding or variable region.
  • antibody fragments include, but are not limited to, F ab , F ab′ , F (ab′)2 and F v fragments.
  • a fragment or analog of an anti-IL-4 and/or IL-13 antibody is one which can prevent or substantially reduce the ability of the receptor to bind to a ligand or to initiate signaling.
  • fragment generally are synonymous and, with respect to antibodies, can refer to fragments, such as F v , F ab , F (ab′)2 and so on which can prevent or substantially reduce the ability of the receptor to bind to a ligand or to initiate signaling.
  • An “F v ” fragment consists of a dimer of one heavy and one light chain variable domain in a non-covalent association (V H -V L dimer).
  • V H -V L dimer the three CDRs of each variable domain interact to define a target binding site on the surface of the V H -V L dimer, as in an intact antibody.
  • the six CDRs confer target binding specificity on the intact antibody.
  • a single variable domain or half of an F v comprising only three CDRs specific for a target
  • Single-chain F v ,” “sF v ” or “scAb” antibody fragments comprise the V H and V L domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the F v polypeptide further comprises a polypeptide linker, often a flexible molecule, between the V H and V L domains, which enables the sFv to form the desired structure for target binding.
  • diabody refers to an antibody fragment with two antigen-binding sites, which can comprise a heavy chain variable domain (V H ) connected to a light chain variable domain (V L ) in the same polypeptide chain.
  • V H heavy chain variable domain
  • V L light chain variable domain
  • F ab contains the variable and constant domains of the light chain and the variable and first constant domain (C H1 ) of the heavy chain.
  • F ab′ fragments differ from F ab fragments by the addition of a few residues at the carboxyl terminus of the C H1 domain to include one or more cysteines from the antibody hinge region.
  • F ab′ fragments can be produced by cleavage of the disulfide bond at the hinge cysteines of the F (ab′)2 pepsin digestion product. Additional enzymatic and chemical treatments of antibodies can yield other functional fragments of interest.
  • linear Fab refers to a tetravalent antibody as described by Miller et al. (2003), J Immunol. 170: 4854-4861.
  • a linear Fab is composed of a tandem of the same CH1-VH domain, paired with the identical light chain at each CH1-VH position.
  • Monoclonal antibodies herein specifically include “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass (type or subtype), with the remainder of the chain(s) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity of binding to IL-4 and/or IL-13 or impacting IL-4 and/or IL-13 activity or metabolism (U.S. Pat. No. 4,816,567; and Morrison et al. (1984), Proc Natl Acad Sci USA 81:6851).
  • CDRs from one class of antibody can be grafted into the FR of an antibody of different class or subclass.
  • Monoclonal antibodies are highly specific, being directed against a single target site, epitope or determinant. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes) of an antigen, each monoclonal antibody is directed against a single determinant on the target. In addition to their specificity, monoclonal antibodies are advantageous being synthesized by a host cell, uncontaminated by other immunoglobulins, and provides for cloning the relevant gene and mRNA encoding the antibody of chains thereof.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies for use with the present invention may be isolated from phage antibody libraries using well known techniques or can be purified from a polyclonal preparation.
  • the parent monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method described by Kohler et al. (1975), Nature 256:495, or may be made by recombinant methods well known in the art.
  • polyvalent antibody refers to an antibody comprising two or more antigen binding sites, thus being able to bind two or more antigens, which may have the same or a different structure, simultaneously.
  • bivalent means that the antibody comprises two antigen binding sites.
  • tetravalent means that the antibody comprises four antigen binding sites.
  • an antigen binding site refers to the part of the antibody which comprises the area which specifically binds to and is complementary to part or all of an antigen. Where an antigen is large, an antibody may only bind to a particular part of the antigen, which part is termed on epitope.
  • An antigen binding domain may be provided by one or more antibody variable domains. Preferably, an antigen binding domain is made of the association of an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (VH).
  • antigen refers to a molecule or a portion of a molecule capable of being bound by the antibodies of the present invention.
  • An antigen can have one or more than one epitope.
  • antigens recognized by the antibodies of the present invention include, but are not limited to, serum proteins, e.g. cytokines such as IL-4, IL-5, IL-9 and IL-13, bioactive peptides, cell surface molecules, e.g. receptors, transporters, ion-channels, viral and bacterial proteins.
  • the term “monospecific” as used in the present invention means that the polyvalent antibody of the present invention recognizes only one antigen, all the antigen binding sites being identical.
  • bispecific means that the polyvalent antibody of the present invention recognizes two different epitopes on the same or on two different antigens.
  • BsAb bispecific antibody
  • a bispecific antibody is able to bind two different antigens simultaneously.
  • BsAbs pave the way for new therapeutic applications by redirecting potent effector systems to diseased areas or by increasing neutralizing or stimulating activities of antibodies.
  • BsAbs bispecific antibodies
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Bispecific antibodies were originally made by fusing two hybridomas, each capable of producing a different immunoglobulin (Milstein and Cuello, 1983, 1984), but the complexity of species (up to ten different species) produced in cell culture makes purification difficult and expensive (George and Huston, 1997).
  • factors made them impractical for large scale therapeutic applications. Such factors include: rapid clearance of heteroconjugates in vivo, the laboratory intensive techniques required for generating either type of molecule, the need for extensive purification of heteroconjugates away from homoconjugates or mono-specific antibodies and generally low yields.
  • An embodiment of the invention is a bispecific antibody that has been engineered to comprise a dual-V-region antibody-like protein or fragment thereof that specifically binds to two different epitopes on the same or on two different antigens.
  • An embodiment of the invention a bispecific antibody or bispecific antibody fragment thereof that specifically binds to IL-13 and IL-4, wherein said bispecific antibody or bispecific antibody fragment thereof comprises a variable light chain domain and a variable heavy chain domain, wherein said variable light chain domain comprises amino acid sequences SEQ ID NO:1 and SEQ ID NO:3.
  • a further embodiment of the invention is a bispecific antibody or bispecific antibody fragment thereof that specifically binds to IL-13 and IL-4, wherein said bispecific antibody or bispecific antibody fragment thereof comprises a variable light chain domain and a variable heavy chain domain, wherein said variable heavy chain domain comprises amino acid sequences SEQ ID NO:2 and SEQ ID NO:5.
  • Another embodiment of the invention is a bispecific antibody or bispecific antibody fragment thereof that specifically binds to IL-13 and IL-4, wherein said bispecific antibody or bispecific antibody fragment thereof comprises a variable light chain domain and a variable heavy chain domain, wherein said variable heavy chain domain comprises amino acid sequences SEQ ID NO:2 and SEQ ID NO:4.
  • An embodiment of the invention is a bispecific antibody or bispecific antibody fragment thereof that specifically binds to IL-13 and IL-4, wherein said bispecific antibody or bispecific antibody fragment thereof comprises a variable light chain domain comprising amino acid sequences SEQ ID NO:1 and SEQ ID NO:3, and a variable heavy chain domain comprising amino acid sequences SEQ ID NO:2 and SEQ ID NO:4.
  • a further embodiment of the invention is a bispecific antibody or bispecific antibody fragment thereof that specifically binds to IL-13 and IL-4, wherein said bispecific antibody or bispecific antibody fragment thereof comprises a variable light chain domain comprising amino acid sequences SEQ ID NO:1 and SEQ ID NO:3, and a variable heavy chain domain comprising amino acid sequences SEQ ID NO:2 and SEQ ID NO:4, wherein a peptide linker links SEQ ID NO:1 to SEQ ID NO:3, and a peptide linker links SEQ ID NO:2 to SEQ ID NO:4.
  • An embodiment of the invention is huTBTI3_2_1 or SAR156597 comprising a bispecific antibody or bispecific antibody fragment thereof that specifically binds to IL-13 and IL-4, comprising (a) variable light chain domain comprising the amino acid sequences of SEQ ID NO:1 and SEQ ID NO:3; (b) a variable heavy chain domain comprising the amino acid sequences of SEQ ID NO:2 and SEQ ID NO:4; (c) a peptide linker linking SEQ ID NO:1 to SEQ ID NO:3, and a peptide linker linking SEQ ID NO:2 to SEQ ID NO:4 wherein the peptide linker has an amino acid sequence consisting of SEQ ID NO:6; and (d) constant region domains.
  • multispecific means that the polyvalent antibody of the present invention recognizes multiple different epitopes on the same or on multiple different antigens.
  • linker refers to a peptide adapted to connect the variable domains of the antibody constructs of the present invention.
  • the peptide linker may contain any amino acids, the amino acids glycine (G) and serine (S) being preferred.
  • the linkers may be equal or differ from each other between and within the heavy chain polypeptide and the light chain polypeptide.
  • the linker may have a length of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acids.
  • a preferred peptide linker unit for the heavy chain domains as for the light chain domains is GGGGS.
  • the numbers of linker units of the heavy chain and of the light chain may be equal (symmetrical order) or differ from each other (asymmetrical order).
  • a peptide linker is preferably long enough to provide an adequate degree of flexibility to prevent the antibody moieties from interfering with each others activity, for example by steric hindrance, to allow for proper protein folding and, if necessary, to allow the antibody molecules to interact with two or more, possibly widely spaced, receptors on the same cell; yet it is preferably short enough to allow the antibody moieties to remain stable in the cell.
  • the length, composition and/or conformation of the peptide linkers can readily be selected by one skilled in the art in order to optimize the desired properties of the polyvalent antibody.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as F v , F ab , F ab′ , F (ab′)2 or other target-binding subsequences of antibodies) which contain sequences derived from non-human immunoglobulin, as compared to a human antibody.
  • the humanized antibody will comprise substantially all of one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin template sequence.
  • the humanized antibody may also comprise at least a portion of an immunoglobulin constant region (F c ), typically that of the human immunoglobulin template chosen.
  • F c immunoglobulin constant region
  • the goal is to have an antibody molecule that is minimally immunogenic in a human.
  • one or more amino acids in one or more CDRs also can be changed to one that is less immunogenic to a human host, without substantially minimizing the specific binding function of the one or more CDRs to IL-4 and/or IL-13.
  • the FR can be non-human but those amino acids most immunogenic are replaced with ones less immunogenic. Nevertheless, CDR grafting, as discussed above, is not the only way to obtain a humanized antibody.
  • modifying just the CDR regions may be insufficient as it is not uncommon for framework residues to have a role in determining the three-dimensional structure of the CDR loops and the overall affinity of the antibody for its ligand.
  • any means can be practiced so that the non-human parent antibody molecule is modified to be one that is less immunogenic to a human, and global sequence identity with a human antibody is not always a necessity.
  • humanization also can be achieved, for example, by the mere substitution of just a few residues, particularly those which are exposed on the antibody molecule and not buried within the molecule, and hence, not readily accessible to the host immune system.
  • Antibody homolog refers to any molecule which specifically binds IL-4 and/or IL-13 as taught herein.
  • an antibody homolog includes native or recombinant antibody, whether modified or not, portions of antibodies that retain the biological properties of interest, such as binding IL-4 or IL-13, such as an F ab or F v molecule, a single chain antibody, a polypeptide carrying one or more CDR regions and so on.
  • the amino acid sequence of the homolog need not be identical to that of the naturally occurring antibody but can be altered or modified to carry substitute amino acids, inserted amino acids, deleted amino acids, amino acids other than the twenty normally found in proteins and so on to obtain a polypeptide with enhanced or other beneficial properties.
  • Antibodies with homologous sequences are those antibodies with amino acid sequences that have sequence homology with the amino acid sequence of a IL-4, IL-13 or bispecific IL-4/IL-13 antibody of the present invention.
  • homology is with the amino acid sequence of the variable regions of an antibody of the present invention.
  • Sequence homology as applied to an amino acid sequence herein is defined as a sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence homology to another amino acid sequence, as determined, for example, by the FASTA search method in accordance with Pearson & Lipman, Proc Natl Acad Sci USA 85, 2444-2448 (1988).
  • a chimeric antibody is one with different portions of an antibody derived from different sources, such as different antibodies, different classes of antibody, different animal species, for example, an antibody having a variable region derived from a murine monoclonal antibody paired with a human immunoglobulin constant region and so on.
  • a humanized antibody is a species of chimeric antibody.
  • Methods for producing chimeric antibodies are known in the art, see, e.g., Morrison, 1985, Science 229:1202; Oi et al., 1986, BioTechniques 4:214; Gillies et al., 1989, J Immunol Methods 125:191-202; and U.S. Pat. Nos. 5,807,715, 4,816,567, and 4,816,397.
  • scFv fragments include scFv fragments, chimeric antibodies, diabodies, triabodies, tetrabodies and molecular recognition units (mrus) (see reviews by Winter & Milstein, 1991, Nature 349:293-299; and Hudson, 1999, Curr Opin Imm 11:548-557), each with antigen-binding or epitope-binding ability.
  • scF v single chain F v fragment
  • the V H and V L domains of an antibody are linked by a flexible peptide.
  • the linker is a peptide of about 15 amino acids. If the linker is much smaller, for example, 5 amino acids, diabodies are formed.
  • the smallest binding unit of an antibody is a CDR, typically the CDR2 of the heavy chain which has sufficient specific recognition and binding capacity.
  • a fragment is called a molecular recognition unit or mru.
  • mru molecular recognition unit
  • Several such mrus can be linked together with short linker peptides, therefore forming an artificial binding protein with higher avidity than a single mru.
  • functional equivalents include antibodies with homologous sequences, antibody homologs, chimeric antibodies, artificial antibodies and modified antibodies, for example, wherein each functional equivalent is defined by the ability to bind to IL-4 and/or IL-13, inhibiting IL-4 and/or IL-13 signaling ability or function, or inhibiting binding of IL-4 and/or IL-13 to its receptor.
  • modified antibodies include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, deamidation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand, linkage to a toxin or cytotoxic moiety or other protein etc.
  • the covalent attachment need not yield an antibody that is immune from generating an anti-idiotypic response.
  • the modifications may be achieved by known techniques, including, but not limited to, specific chemical cleavage, acetylation, formylation, metabolic synthesis etc.
  • the modified antibodies may contain one or more non-classical amino acids.
  • “Mammal” for purposes of treatment refers to any animal classified as a mammal, including human, domestic and farm animals, nonhuman primates, and zoo, sports or pet animals, such as dogs, horses, cats, cows etc.
  • treatment refers to both therapeutic treatment and prophylactic or preventative measures as a course of therapy. It refers to preventing, curing, reversing, attenuating, alleviating, minimizing, suppressing or halting deleterious effects of a disease state, disease progression, disease causative agent (e.g., bacteria or viruses) or other abnormal condition.
  • disease causative agent e.g., bacteria or viruses
  • an “isolated” or “purified” antibody is substantially free of cellular material or other contaminating proteins from the cell or tissue source or medium from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • the language “substantially free of cellular material” includes preparations of an antibody in which the polypeptide/protein is separated from cellular components of the cells from which same is isolated or recombinantly produced.
  • an antibody that is substantially free of cellular material includes preparations of the antibody having less than about 30%, 20%, 10%, 5%, 2.5% or 1%, (by dry weight) of contaminating protein.
  • the antibody When the antibody is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, 10%, 5%, 2.5% or 1% of the volume of the protein preparation.
  • culture medium represents less than about 20%, 10%, 5%, 2.5% or 1% of the volume of the protein preparation.
  • antibody When antibody is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals and reagents, i.e., the antibody of interest is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly, such preparations of the antibody have less than about 30%, 20%, 10%, 5% or 1% (by dry weight) of chemical precursors or compounds other than antibody of interest.
  • antibodies are isolated or purified.
  • therapeutic agent and “therapeutic agents” refer to any agent(s) which can be used in the treatment, management or amelioration of a disease, disorder, malady and the like associated with aberrant IL-4 and/or IL-13 metabolism and activity.
  • dose refers to the quantity of any agent(s) which can be used in the treatment, management or amelioration of a disease, disorder, malady and the like associated with aberrant IL-4 and/or IL-13 metabolism and activity.
  • safe dose refers to any agent(s) or dose of any agent(s) which can be used in the treatment, management or amelioration of a disease, disorder, malady and the like associated with aberrant IL-4 and/or IL-13 metabolism and activity while maintaining a clinically acceptable benefit/risk profile.
  • a safe dose of the dual-V-region antibody-like binding proteins or fragments thereof disclosed herein is selected from the group consisting of 10 mg, 20 mg, 40 mg, 50 mg, 80 mg, 100 mg, 150 mg, 200 mg, and 300 mg.
  • An embodiment of a safe dose is about 10 mg to about 300 mg.
  • a further embodiment of a safe dose of a is any dose that is 200 mg, about 200 mg, up to 200 mg, or no greater than about 200 mg.
  • a safe dose is about 50 mg, or about 100 mg, or about 200 mg.
  • the safe dose is administered once weekly.
  • the safe dose is administered subcutaneously (SC).
  • Intracellular signaling after ligation of IL-4 and IL-13 with their cell surface receptors is mediated in part by phosphorylation of the signaling molecule signal transducer and activator of transcription 6 (Stat6).
  • Chemokine (C-C motif) ligand 17 is a small cytokine belonging to the CC chemokine family.
  • CCL17 is also known as thymus and activation regulated chemokine (TARC).
  • TARC is induced by IL-4 and/or IL-13 through Stat6 phosphorylation (Wirnsberger et al., (2006) Eur J Immunol. 36: 1882-91; Liddiard et al., (2006) BMC Mol Biol. 29: 7:45; Monick et al., (2007) J Immunol.
  • the methods disclosed herein comprise methods of detecting the binding to IL-4 and/or IL-13 of an antibody or antibody-like binding protein or fragment thereof that has been administered to a subject, the methods comprising (a) administering the antibody or antibody-like binding protein of fragment thereof to the subject; and (b) determining the amount of CCL17/TARC within a blood, serum, or plasma sample drawn from the subject, wherein a decrease in the amount of CCL17/TARC in the sample relative to a sample drawn from the subject prior to administration of the antibody or antibody-like binding protein or fragment thereof signifies binding of the antibody or antibody-like binding protein or fragment thereof to IL-4 and/or IL-13.
  • step (c) further comprises increasing the dose if the decrease in TARC/CCL17 measured in step (b) is below a threshold value (i.e. if TARC/CCL17 levels do not decrease enough), or decreasing the dose if the decrease in TARC/CCL17 measured in step (b) is above a threshold value (i.e. if TARC/CCL17 decreases too much).
  • the threshold value of step (c) is about a 10% decrease, or about a 15% decrease, or about a 20% decrease, or about a 25% decrease, or about a 30% decrease, or about a 35% decrease, or about a 40% decrease, or about a 45% decrease, or about a 50% decrease, or about a 55% decrease, or about a 60% decrease, or about a 65% decrease in the amount of TARC/CCL17 relative to the amount of TARC/CCL17 in the subject measured before the dose was administered.
  • the threshold value is about a 20% to about a 60% decrease, or about a 40% to about a 50% decrease in the amount of TARC/CCL17 relative to the amount of TARC/CCL17 in the subject measured before the dose was administered. In some embodiments, the threshold value is about a 43% decrease in the amount of TARC/CCL17 relative to the amount of TARC/CCL17 in the subject measured before the dose was administered. For example, a 43% decrease for a 200 mg dose signifies binding of a 200 mg dose of bispecific anti-IL-4/IL-13 dual-V-region antibody-like binding protein to IL-4/IL-13.
  • huTBTI3_2_1 and “SAR156597” are interchangeable and refer to the same dual-V-region antibody-like protein comprising a variable light chain comprising amino acid sequences SEQ ID NO:1 and SEQ ID NO:3 and a variable heavy chain comprising amino acid sequences SEQ ID NO:2 and SEQ ID NO:4.
  • IPF idiopathic pulmonary fibrosis
  • each dose cohort 8 patients (6 receiving SAR156597 and 2 receiving placebo) received repeated SC, once-weekly doses of SAR156597 or matching placebo control.
  • the second cohort was initiated after the review of the safety of the first cohort by the DMC.
  • the third cohort at a dose of 200 mg was initiated after the review of the safety of the 2 preceding cohorts.
  • the study duration was 22 weeks, as follows: 4 weeks of screening; 6 weeks of treatment period (7 administrations); 12 weeks follow-up.
  • ADAs anti-drug antibodies
  • the investigational medicinal product was provided as SAR156597 in lyophilized form for the preparation of SC dose solution.
  • Each vial containing 185 mg of SAR156597 plus excipients was stored between 2° C. and 8° C. (36° F. and 46° F.).
  • the IMP was reconstituted on the morning of dosing (no more than 1 hour prior to SC injection) with 1.7 mL sterile, nonpyrogenic distilled water at room temperature.
  • concentrations of the constituents in solution after reconstitution for injection were: 100 mg/mL of SAR156597 in 6.3 mmol/L monobasic sodium phosphate, 3.7 mmol/L tromethamine, 5% (weight/volume) sucrose, 3% (w/V) proline, and 0.2% (w/V) polysorbate 80 with a final pH of 7.0.
  • each vial containing 2 mL of liquid containing the same excipients at the same concentrations as for the reconstituted SAR156597 formulation was provided.
  • the IMP (SAR156597 or placebo) was administered as periumbilical SC injections. Within 1 hour of reconstitution, the dose was administered in a zone 4 to 10 cm from the umbilicus in a left or right quadrant above the waistline.
  • SAR156597 is an engineered humanized bi-specific immunoglobin G (IgG)-4 antibody that binds and neutralizes both IL-4 and IL-13. SAR156597 shows high affinity for IL-4 and IL-13 from both humans and cynomolgus monkeys.
  • IgG immunoglobin G
  • Pulmonary function test Carbon monoxide diffusing capacity, corrected for hemoglobin (DLCO); Forced (expiratory) vital capacity (FVC); Forced expiratory volume over 1 second (FEV1); Total lung capacity (body plethysmography) (TLC); Residual volume (body plethysmography) (RV).
  • DLCO hemoglobin
  • FVC Forced (expiratory) vital capacity
  • FEV1 Forced expiratory volume over 1 second
  • TLC Total lung capacity
  • RV Residual volume
  • Two PFTs were performed within 3 weeks prior to dosing. The first PFT was performed to screen the patient and the data from the second PFT (at the randomization visit) was averaged with the first one to establish a baseline value; these tests were performed on separate days.
  • a PFT was also performed at Visit 8 (Week 6)/end-of-treatment (EOT) and Visit 14 (Week 18/12 weeks following the last dose of study treatment).
  • Oxygen saturation was assessed using SpO2 at screening, baseline, at Week 6/EOT, and at Week 18 (12 weeks following the last dose of study treatment). The first SpO2 measurement was performed at the screening and the data from the second measurement was averaged with the first one to establish a baseline value; these tests were performed on separate days.
  • TARC was quantified using a human CCL17/TARC Quantikine ELISA kit from R&D Systems, following the instructions of the kit manual. For this assay, the lower limit of quantification was 31.2 pg/mL and the upper limit of quantification was 2000 pg/mL.
  • Protein biomarkers included chemokine [C-C motif] ligand 18 [CCL-18], Krebs von den Lundgen-6 [KL-6], surfactant protein A [SP-A], surfactant protein D [SP-D], mammalian chitinase-like proteins [YKL-40], IL-4, IL-13, IL-8, immunoglobin E [IgE], eotaxin, inter-cellular adhesion molecule 1 [ICAM1], periostin, thymus and activation-regulated chemokine [TARC] CCL-17, matrix metalloproteinase-7 [MMP7]), and RNA (mRNA, micro-RNA) expressions.
  • PD endpoints The changes from baseline in the 5 PFTs (FVC, FEV1, TLC, DLCO, and RV) were analyzed. At the end of treatment (EOT, at Visit 8, Week 6), the patient lung function was overall not altered as evident by the mean change from baseline for the main parameters (percent predicted FVC and percent predicted DLCO) which was comparable between treatment groups. Twelve weeks after the EOT, the mean change from baseline for % predicted FVC and % predicted DLCO showed that lung function remained unchanged in all treatment groups.
  • Secondary PD endpoints Secondary PD variables (SpO2, protein/RNA biomarkers, and the SGRQ) were measured at the end of the 6-week treatment and post-treatment follow-up periods. These PD results were inconclusive due to the small sample size of the study and the short treatment duration.
  • Lipasemia ⁇ 3 ULN Amylasemia ⁇ 3 ULN Glucose Hypoglycaemia ⁇ 3.9 mmol/L and ⁇ LLN ADA May 2005.
  • Hematocrit ⁇ 0.37 v/v (Male); ⁇ 0.32 v/v (Female) ⁇ 0.55 v/v (Male); ⁇ 0.5 v/v (Female) RBC ⁇ 6 Tera/L Unless specifically required for particular drug development, the analysis is redundant with that of Hb. Otherwise, consider FDA criteria. Platelets ⁇ 100 Giga/L International Consensus meeting ⁇ 700 Giga/L on drug-induced blood cytopenias, 1991. Urinalysis pH ⁇ 4.6 ⁇ 8 Vital signs HR ⁇ 50 bpm and decrease from baseline ⁇ 20 bpm To be applied for all positions ⁇ 120 bpm and increase from baseline ⁇ 20 bpm (including missing) except STANDING.
  • Prolonged* Prolonged >450 ms (Male); >470 ms (Female) Additional ⁇ 500 ms Increase from baseline Borderline: Increase from baseline 30-60 ms
  • Prolonged Increase from baseline >60 ms *QTc prolonged and ⁇ QTc > 60 ms are the PCSA to be identified in individual subjects/patients listings.
  • AEs adverse events
  • Pre-treatment adverse events AEs that occurred or worsened during the pre-treatment phase
  • Treatment-emergent adverse events AEs that occurred or worsened during the on-treatment phase
  • Post-treatment adverse events AEs that occurred or worsened during the post-treatment phase.
  • TEAEs were assigned to the IMP received at the time of the AE onset.
  • TEAEs were summarized and listed by primary system organ class (SOC) and preferred term (PT). In addition, all AEs were listed, sorted by patient and onset date and time.
  • SOC primary system organ class
  • PT preferred term
  • the baseline value in this study was the value collected during Day 1 predose assessments.
  • the body weight was analyzed as the raw parameter value and the percent change from baseline. Individual body mass index was analyzed as raw value.
  • the baseline values were the predose values collected on the Day 1. For all parameters, an “on-treatment” analysis was performed using all postbaseline values collected during the on-treatment phase, including all rechecked values.
  • Heart rate and blood pressure were analyzed as raw parameter value (for supine and standing position available), the change from baseline (for supine position only), and as orthostatic parameter (standing-supine parameter values).
  • the baseline values were the predose values collected on Day 1.
  • an “on-treatment” analysis was performed using all postbaseline values collected during the on-treatment phase, including all unplanned and rechecked values.
  • the body temperature was analyzed as raw parameter value and change from baseline.
  • the baseline values were the predose values collected on the Day 1.
  • Heart rate, PR-, QRS-, QT-, and corrected QT-interval (QTc) were analyzed as the raw parameter value and the change from baseline.
  • the baseline values were the predose values collected on Day 1.
  • an “on-treatment” analysis was performed using all postbaseline values collected during the on-treatment phase, including all rechecked values.
  • Erythema size and edema size were summarized in descriptive statistics by parameter, treatment group, and time-point.
  • the numbers (%) of patients with the most extreme qualitative assessment of present pain intensity, erythema, edema, itch, papule, vesiculation, and pustule grades were summarized over the entire study by treatment group.
  • TEAEs treatment-emergent adverse events
  • the number of patients with QTcB prolongations was higher in the treatment groups compared to the placebo group. There were 4 patients (1 patient in the SAR156597 50 mg group, 2 patients in the SAR156597 100 mg group, and 1 patient in the SAR156597 200 mg group) who had QTcB prolongations during this study.
  • the number of patients with at least 1 TEAE was comparable among the treatment groups (5 of 6 patients in the placebo and 200 mg groups, 6 of 6 patients in the 50 mg and 100 mg groups), most TEAEs were mild to moderate in intensity.
  • the most common reported TEAE was infections, which was balanced among the treatment groups (3 of 6 patients in the placebo, 50 mg, and 200 mg groups, 2 of 6 patients in 100 mg group).
  • SAE Serious adverse event
  • Samples for PK analysis were collected pre-dose (within 2 hours before each dose administration) during Visits 2 to 8.
  • Pharmacokinetics samples during Visits 9/early termination (ET), 11, and 14 were collected in the morning.
  • Anti-SAR156597 antibodies (ADA) samples were collected at approximately the same times as the PK samples at Visits 2, 9/ET, 11, and 14. If, at Week 12, ADA related to immunogenicity were present, an additional follow-up visit occurred at a minimum of 6 months after the first dose to examine the elevated parameter.
  • SAR156597 plasma concentrations were determined using a validated enzyme linked immunosorbent assay (ELISA) method with a lower limit of quantification (LLOQ) of 0.05 ⁇ g/mL (DOH0850) under the responsibility of Bertin Pharma (L.E.M.M. [Laboratoire d'Etude du Métabolisme des Médicaments], DSV/iBiTec-S/SPI, CEA-Saclay, Gif sur Yvette Cedex, France). All raw data from the bioanalytical studies are stored at Bertin Pharma according to the procedures in use at the test site.
  • ELISA enzyme linked immunosorbent assay
  • ADA in plasma was assayed using a validated ELISA method (DOH0851) under the responsibility of Disposition, Safety & Animal Research Operational Center of adjoin (Biomarker and Biological Assays group), Sanofi. All samples were first evaluated using a screening assay. Samples found positive in the screening assay were then tested in a confirmatory assay. A titer was reported only for samples confirmed to be positive. All raw data from the bioanalytical studies are stored at sanofi-aventis, adjoin, France, according to the procedures in use at the test site.
  • Pharmacokinetic parameters were estimated using the population PK (Bayesian) approach and are presented in Table 5.
  • Bayesian analysis was performed with the NONMEM® computer program (version 7.1.2) running on a LINUX cluster of multi-processor computers.
  • a Bayesian Data Set was constructed using data of the 3 cohorts (doses 50, 100, and 200 mg).
  • the NONMEM software version 7.1.2 was used to analyze the data.
  • the final population PK model obtained in POH0338 study (constructed using PK data from an earlier study) was applied to the Bayesian Data Set, with its parameter estimates as prior estimates for the assessment of individual parameters and concentration predictions.
  • C max , AUC 0-168h , and t 1/2z were log-transformed.
  • the plasma drug concentration lower than the LLOQ of 0.05 ⁇ g/mL for SAR156597 was treated as zero in calculating the mean values. Means and their associated statistics were generated from unrounded numbers and may differ slightly from those values which would have been determined using rounded numbers. Once final PK analysis was performed, the PK parameters were transferred electronically to the Biostatistics Department for further statistical analysis. Concentrations and PK parameter values were rounded to 3 significant figures.
  • SAR156597 trough concentrations are graphically presented in FIG. 4 .
  • Bayesian PK analysis predicted that the steady state for AUC 0-168ss was reached at 89% for 50 mg dose group, 94.8% for 100 mg dose group, and 93.7% for 200 mg dose group after seventh dose of SAR156597 on Week 6.
  • SAR156597 exposure increased slightly more than dose proportionally.
  • a 4-fold increase in SAR156597 dose demonstrated a 5.21- to 5.92-fold increase in C max and a 5.17- to 5.83-fold increase in AUC 0-168 .
  • ADAs Two patients had ADA positive samples.
  • Bayesian PK analysis predicted that 89% to 94.7% of steady state was reached after 7 doses of SAR156597 on Week 6.
  • SAR156597 exposure increased slightly more than dose proportionally.
  • a 4-fold increase in SAR156597 dose demonstrated a 5.21- to 5.92-fold increase in C max and a 5.17- to 5.83-fold increase in AUC 0-168 .
  • Example 5 IL-13 is a Driver of Pulmonary Fibrosis in a FRA-2 Transgenic Mouse Model
  • Interstitial lung disease refers to a large group of more than 200 lung diseases affecting the interstitium.
  • a significant subset of scleroderma patients show pulmonary manifestation with parenchymal lung involvement resulting in interstitial lung abnormalities and compromised pulmonary function. This fatal end-stage condition is characterized by interstitial pneumonia and scarring.
  • the AP-1 family of transcription factors regulates the expression of a number of target genes that control a variety of cellular functions.
  • the AP-1 complex is composed of Jun and Fos proteins.
  • Fos-Like Antigen 2, FRA-2 a member of the Fos family of proteins, participates in the AP-1 complex formation with a regulatory function.
  • This example describes the characterization of a new line of transgenic mice overexpressing the Fra-2 gene. These mice showed fibrosis of the lungs during development starting at thirteen weeks. The development of fibrosis coincides with elevated levels of circulating and pulmonary Th2 cytokines.
  • an EGFP gene was engineered into the construct to enable visualization of the transgene expression.
  • the full length FRA2 gene was driven by an H2kb promoter resulting in the ubiquitous expression of the transgene.
  • the Eferl transgenic construct is shown in FIG. 19A and consists of an H2Kb promoter, the genomic Fra-2 locus, a reporter IRES-EGFP sequence, and a LTR sequence harboring a polyadenylation signal (pA).
  • E1-E4 are Exons 1-4 of Fra-2; HindIII (H) restriction sites and probe location (P) used for Southern blot analysis are indicated.
  • FIG. 19B shows a schematic of the transgenic vector used here, which contains a mouse H2Kb promoter driving mouse Fra-2 gene and a T2A-EGFP-polyA-loxP-hUBp-EM7-Neo-loxP cassette (4,898 bp).
  • Four to eight copies of the transgene were randomly integrated into the host chromosome.
  • Five founder transgenic lines over-expressing FRA2 were generated; three lines were triaged for further characterization based on transgene expression.
  • the genomic coordinates of the fragments were as follows: H2Kb (H2-K1) promoter: Chr17: 34,137,222-34,139,244 ( ⁇ ) after Morello et al., 1986 , EMBO J. 5(8): 1877-83; Fra-2 (Fosl2) gene (without stop): Chr5: 32,438,859-32,455,559.
  • the FRA-2 over-expressing transgenic mice exhibited increasing lung weights with age accompanied by an increased deposition of collagen measured by hydroxyproline content ( FIG. 5 ).
  • FIG. 6 Histological analysis also showed increased deposition of collagen with time in lung samples and was conducted as follows. Following euthanasia, the lungs were insufflated with fixative under a fixed pressure. The lungs were then removed and placed into fixative. After fixation was complete, the lungs were embedded into a 1% agar solution and were sliced transsagitally into 3 mm thick step sections. After processing, all sections were embedded into paraffin blocks. All paraffin blocks were microtomed at 4 ⁇ m and stained using Masson's Trichrome stain to demonstrate collagen deposition (blue) ad H&E stain to show overall morphology ( FIG. 6 ).
  • IL-4, IL-5 and IL-13 Th2 cytokine profile
  • This developmental pulmonary fibrosis mouse model was used to study a role for IL-4 and IL-13 in fibrosis.
  • a role for IL-13 in promoting fibrosis was evaluated using a validated surrogate mouse IL-13 antibody (since there is no cross reactivity of SAR156597 in rodents) with neutralizing activity in the FRA2 mouse model.
  • Thirteen week old FRA2 mice were dosed with the surrogate mouse IL-13 antibody, at 10 mpk, administered by the ip route twice a week for 29 days.
  • a rat IgG1 isotype at 10 mpk was dosed in parallel FRA2 mice as controls.
  • a third group of FRA2 mice received saline as a second control group.
  • the below table shows the different treatment groups used in the study.
  • mice were euthanized and their lungs removed and processed to provide (1) lung homogenates for protein analysis, (2) RNA preparation, and (3) one lung lobe was tied off and insufflated, excised and fixed for histological analysis.
  • the lung homogenates were used to quantitate the amount of collagen.
  • FIG. 11 the hydroxyproline content of FRA2 mouse lungs was increased compared to the wt littermate controls.
  • the hydroxyproline levels in the IL-13 Ab treated FRA2 mouse lungs was significantly decreased.
  • Transcript analysis by real time PCR was used to follow the expression of a variety of fibrotic markers, profibrotic mediators and genes that are regulated by IL-13.
  • Results for IPF biomarkers FN1, SPDEF, and MUC5B are shown in FIG. 14 .
  • Results for profibrotic markers CCL2 and CCL11 are shown in FIG. 15 .
  • Results for IL-6, ARG1, and IL13Ra2 are shown in FIG. 16 .
  • IL-13-regulated protein expression was further studied in lung homogenates by ELISA.
  • IL-13, IL-4 and IL-17 levels were increased in the FRA2 lung homogenates compared to wild type lungs and these cytokine levels were attenuated following anti-IL-13 Ab treatment ( FIG. 17 ).
  • Decreased IL-13 levels confirmed antibody loading in the lungs following the 4 wk treatment ( FIG. 17 ).
  • MCP-1(CCL2) is a well characterized chemokine associated with pulmonary fibrosis and is regulated by IL-13.
  • CCL17/TARC and YKL-40 are IL-13 regulated proteins. Following the surrogate Ab treatment, CCL2, CCL17 and YKL-40 were significantly inhibited ( FIG. 18 ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
US15/320,106 2014-06-27 2015-06-26 Anti-il4-il 13 bispecific antibodies Abandoned US20170145089A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/320,106 US20170145089A1 (en) 2014-06-27 2015-06-26 Anti-il4-il 13 bispecific antibodies

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201462018253P 2014-06-27 2014-06-27
EP14306477 2014-09-24
EP14306477.2 2014-09-24
US201562102097P 2015-01-11 2015-01-11
US201562102555P 2015-01-12 2015-01-12
PCT/IB2015/001377 WO2015198146A2 (en) 2014-06-27 2015-06-26 Anti-il4-il 13 bispecific antibodies
US15/320,106 US20170145089A1 (en) 2014-06-27 2015-06-26 Anti-il4-il 13 bispecific antibodies

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2015/001377 A-371-Of-International WO2015198146A2 (en) 2014-06-27 2015-06-26 Anti-il4-il 13 bispecific antibodies

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/297,419 Continuation US11136388B2 (en) 2014-06-27 2019-03-08 Biomarkers for anti-IL4-IL13 bispecific antibodies

Publications (1)

Publication Number Publication Date
US20170145089A1 true US20170145089A1 (en) 2017-05-25

Family

ID=51663114

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/320,106 Abandoned US20170145089A1 (en) 2014-06-27 2015-06-26 Anti-il4-il 13 bispecific antibodies
US16/297,419 Active US11136388B2 (en) 2014-06-27 2019-03-08 Biomarkers for anti-IL4-IL13 bispecific antibodies

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/297,419 Active US11136388B2 (en) 2014-06-27 2019-03-08 Biomarkers for anti-IL4-IL13 bispecific antibodies

Country Status (18)

Country Link
US (2) US20170145089A1 (zh)
EP (1) EP3160502A2 (zh)
JP (1) JP2017530089A (zh)
KR (1) KR102558891B1 (zh)
CN (1) CN106573975A (zh)
AU (1) AU2015278829B2 (zh)
BR (1) BR112016030568B8 (zh)
CA (1) CA2952902C (zh)
CL (1) CL2016003317A1 (zh)
CR (1) CR20170022A (zh)
EA (1) EA201692495A8 (zh)
MX (1) MX2017000171A (zh)
NZ (1) NZ728020A (zh)
PH (1) PH12016502569A1 (zh)
SG (2) SG10201811562YA (zh)
TN (1) TN2016000569A1 (zh)
TW (2) TWI745962B (zh)
WO (1) WO2015198146A2 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11136388B2 (en) 2014-06-27 2021-10-05 Sanofi Biomarkers for anti-IL4-IL13 bispecific antibodies
US11827671B2 (en) 2019-05-24 2023-11-28 Sanofi Methods for treating systemic sclerosis

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2050764A1 (en) * 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
WO2023166420A1 (en) * 2022-03-03 2023-09-07 Pfizer Inc. Multispecific antibodies and uses thereof

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
IL89489A0 (en) 1988-03-09 1989-09-10 Hybritech Inc Chimeric antibodies directed against human carcinoembryonic antigen
WO1989009622A1 (en) 1988-04-15 1989-10-19 Protein Design Labs, Inc. Il-2 receptor-specific chimeric antibodies
ATE119198T1 (de) 1988-04-16 1995-03-15 Celltech Ltd Verfahren zur herstellung von proteinen mittels rekombinanter dna.
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
WO1993021319A1 (en) 1992-04-08 1993-10-28 Cetus Oncology Corporation HUMANIZED C-erbB-2 SPECIFIC ANTIBODIES
US5989830A (en) 1995-10-16 1999-11-23 Unilever Patent Holdings Bv Bifunctional or bivalent antibody fragment analogue
US20040023338A1 (en) 2001-10-26 2004-02-05 Heavner George A. IL-4 mutein proteins, antibodies, compositions, methods and uses
CA2464695A1 (en) 2001-10-26 2003-05-01 Centocor, Inc. Il-13 mutein proteins, antibodies, compositions, methods and uses
US20060148009A1 (en) 2004-10-12 2006-07-06 Xencor, Inc. Prediction and assessment of immunogenicity
WO2006055638A2 (en) 2004-11-17 2006-05-26 Abgenix, Inc. Fully human monoclonal antibodies to il-13
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
JP2008087194A (ja) 2006-09-29 2008-04-17 Brother Ind Ltd インクジェットプリンタ
EP2050764A1 (en) * 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
JP5561689B2 (ja) * 2007-11-28 2014-07-30 独立行政法人理化学研究所 Il−17rb陽性nkt細胞を用いたアレルギー性気道炎症及び/又は気道過敏症の治療薬のスクリーニング方法
JP5439915B2 (ja) 2009-04-09 2014-03-12 ユニマテック株式会社 硬化性含フッ素ポリエーテル組成物
CA2759848C (en) * 2009-05-05 2018-12-04 Novimmune S.A. Anti-il-17f antibodies and methods of use thereof
WO2011052598A1 (ja) 2009-10-26 2011-05-05 国立大学法人大阪大学 画像特徴抽出装置、画像特徴抽出方法、画像認識装置、及び画像認識方法
WO2011054893A2 (en) * 2009-11-05 2011-05-12 Novartis Ag Biomarkers predictive of progression of fibrosis
US20120156194A1 (en) 2010-12-16 2012-06-21 Genentech, Inc. Diagnosis and treatments relating to th2 inhibition
EP3235508B1 (en) 2011-03-16 2020-12-30 Sanofi Compositions comprising a dual v region antibody-like protein
PL3470432T3 (pl) * 2012-08-21 2022-02-07 Sanofi Biotechnology Sposoby leczenia lub zapobiegania astmie przez podawanie antagonisty IL-4R
RU2698907C2 (ru) * 2012-09-07 2019-09-02 Ридженерон Фармасьютикалз, Инк. Способы лечения атопического дерматита с помощью антагониста il-4r
TWI679019B (zh) 2013-04-29 2019-12-11 法商賽諾菲公司 抗il-4/抗il-13之雙特異性抗體調配物
US20150225479A1 (en) 2014-02-12 2015-08-13 Sanofi Anti-IL-4/Anti-IL-13 Bispecific Antibody/Polyglutamate Formulations
TWI745962B (zh) 2014-06-27 2021-11-11 法商賽諾菲公司 測定投予至人類個體之包括雙-v-區類抗體蛋白或其片段的劑量是否在人類個體中與il-4或il-13特異性結合之方法

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11136388B2 (en) 2014-06-27 2021-10-05 Sanofi Biomarkers for anti-IL4-IL13 bispecific antibodies
US11827671B2 (en) 2019-05-24 2023-11-28 Sanofi Methods for treating systemic sclerosis

Also Published As

Publication number Publication date
PH12016502569A1 (en) 2017-04-17
CL2016003317A1 (es) 2017-08-11
BR112016030568B1 (pt) 2023-10-17
US11136388B2 (en) 2021-10-05
AU2015278829A1 (en) 2017-02-02
JP2017530089A (ja) 2017-10-12
EP3160502A2 (en) 2017-05-03
CN106573975A (zh) 2017-04-19
BR112016030568B8 (pt) 2023-12-05
TWI745962B (zh) 2021-11-11
KR102558891B1 (ko) 2023-07-21
TW201628647A (zh) 2016-08-16
CR20170022A (es) 2017-04-04
NZ728020A (en) 2022-07-01
AU2015278829B2 (en) 2021-03-25
EA201692495A8 (ru) 2017-07-31
EA201692495A1 (ru) 2017-05-31
WO2015198146A3 (en) 2016-03-31
SG11201610594WA (en) 2017-01-27
CA2952902C (en) 2024-02-13
WO2015198146A2 (en) 2015-12-30
SG10201811562YA (en) 2019-01-30
CA2952902A1 (en) 2015-12-30
KR20170020519A (ko) 2017-02-22
BR112016030568A2 (pt) 2018-02-20
MX2017000171A (es) 2017-04-25
TN2016000569A1 (en) 2018-04-04
TW202034954A (zh) 2020-10-01
US20190359703A1 (en) 2019-11-28

Similar Documents

Publication Publication Date Title
US11136388B2 (en) Biomarkers for anti-IL4-IL13 bispecific antibodies
US20220106392A1 (en) Albumin binding domain fusion proteins
CN107660150B (zh) Il-18结合蛋白(il-18bp)和抗体在炎性疾病中
US20210221882A1 (en) Uses of a dual v region antibody-like protein
AU2012252153B2 (en) Anti-nerve growth factor antibodies and methods of preparing and using the same
JP2019510739A (ja) Gfral受容体療法
EP2282757B1 (en) Neutralization of gm-csf for the treatment of heart failure
CN115803057A (zh) 治疗多发性骨髓瘤的方法
US9067989B2 (en) Eotaxin-2 (CCL24) inhibitors in inflammatory, autoimmune, and cardiovascular disorders
JP2016512026A (ja) Pan−ELR+CXCケモカイン抗体
US11827671B2 (en) Methods for treating systemic sclerosis
IL272676A (en) Biospecific antibodies against IL-4 and IL-13
KR20200010294A (ko) 치료용 항-cd40 리간드 항체
WO2013103783A1 (en) Murine il-13 antibodies
US20230272087A1 (en) Method of treating or preventing acute respiratory distress syndrome
US20230391863A1 (en) Bifunctional antagonists of activin and tumor necrosis factor alpha and uses thereof
US20230391880A1 (en) Bifunctional antagonists of activin/tgf-beta and rankl and uses thereof
US20230416381A1 (en) Methods for treating or preventing acute respiratory distress syndrome
US20230391892A1 (en) Bifunctional antagonists of tumor necrosis factor alpha and transforming growth factor beta and uses thereof
KR20170098941A (ko) 인간 dpp-4를 검출하기 위한 검정법

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANOFI, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SOUBRANE, CHRISTINA;JAGERSCHMIDT, ALEXANDRE;SUBRAMANIAM, ARUN;AND OTHERS;SIGNING DATES FROM 20150325 TO 20150407;REEL/FRAME:044739/0712

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION