US20120093833A1 - Human Oncostatin M Antibodies and Methods of Use - Google Patents

Human Oncostatin M Antibodies and Methods of Use Download PDF

Info

Publication number
US20120093833A1
US20120093833A1 US13/269,976 US201113269976A US2012093833A1 US 20120093833 A1 US20120093833 A1 US 20120093833A1 US 201113269976 A US201113269976 A US 201113269976A US 2012093833 A1 US2012093833 A1 US 2012093833A1
Authority
US
United States
Prior art keywords
antibody
seq
amino acid
acid sequence
osm
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
US13/269,976
Other languages
English (en)
Inventor
Juan Carlos Almagro
William DuBell
Johan Fransson
Jose Pardinas
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US13/269,976 priority Critical patent/US20120093833A1/en
Publication of US20120093833A1 publication Critical patent/US20120093833A1/en
Priority to US14/104,520 priority patent/US9163083B2/en
Priority to US14/870,774 priority patent/US9587018B2/en
Priority to US15/423,189 priority patent/US10179812B2/en
Priority to US16/214,891 priority patent/US10941197B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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]
    • C07K16/248IL-6
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • C07K2317/72Increased effector function due to an Fc-modification
    • 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
    • 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

Definitions

  • the present invention relates to a human antibody capable of neutralizing the biological activity produced by oncostatin M binding to membrane receptors on human cells, and uses.
  • Oncostatin M is a 28 kDa multifunctional member of the IL-6 family of cytokines secreted by monocytes, macrophages, neutrophils and activated T-lymphocytes (Tanaka & Miyajima, Rev Physiol Biochem Pharmacol 149: 39-53, 2003). Proteolytic cleavage near the carboxy-terminus of the secreted OSM yields the fully active form of OSM, 209 amino acids length having two N-linked glycosylation sites.
  • OSM belongs to the IL-6 family of cytokines that includes (IL-6, IL-11, leukemia inhibitory factor (LIF), cardiotrophin-1, ciliary neutotrophic factor (CNTF) and cardiotrophin-like cytokine (CLC)) which share a common receptor subunit, gp130 protein.
  • LIF leukemia inhibitory factor
  • CNTF ciliary neutotrophic factor
  • CLC cardiotrophin-like cytokine
  • OSM signals through receptor heterodimers consisting of gp130 and the LIFR ⁇ subunit or gp130 and the OSMR ⁇ subunit.
  • OSM binds gp130 directly and in the absence of any additional membrane-bound co-receptor (Gearing et al., Science 255: 1434-1437, 1992).
  • OSMR ⁇ or LIFR ⁇ are recruited to form a high-affinity signaling complex (Mosley et al., J Biol Chem 271: 32635-32643, 1996). Activation of either receptor results in signaling via the JAK/STAT pathway (Marche et al., J Biol Chem 272: 15760-15764, 1997).
  • OSM is produced primarily by cells of immune system origin and, because of the widespread distribution of its signaling receptors, it has been associated with a variety of biological activities, including cell growth regulation, neural development and regulation of extracellular matrix composition.
  • the present invention provides OSM binding, monoclonal antibodies capable of blocking activities associated with one or more bioactivities associated with OSM and OSM binding receptor interaction on cells, tissues, or organs in a host subject.
  • Amino acid sequences of exemplary OSM binding monoclonal antibodies are provided which can be encoded by nucleic acids for expression in a host cell.
  • One or more of the OSM monoclonal antibodies of the invention define an epitope on the surface of OSM which, when engaged by an antibody of the invention, is prevented from interaction with the receptor components of the signaling complex, gp130 and LIFR ⁇ or gp130 and OSMR ⁇ , thereby preventing ligand ligation driven signaling and downstream biological activity.
  • One aspect of the invention is an isolated antibody reactive with human OSM protein having the antigen binding ability of a monoclonal antibody comprising an antigen binding domain comprising amino acid sequences as set forth in SEQ ID NOs: 13-18 alone or at specified positions of FR1-CDR1-FR2-CDR2-FR3 as set forth in SEQ ID NOs: 1-3, a CDR3 as represented by SEQ ID NO: 27-29 and 47; or an antigen binding domain comprising amino acid sequences as set forth in SEQ ID NOs: 23-26 alone or at specified positions as set forth in SEQ ID NOs: 5-8 and variants thereof, and a CDR3 as represented by SEQ ID NO: 19-22.
  • the human OSM binding antibody comprises a variable domain selected from SEQ ID NO: 49-55.
  • the monoclonal antibody binding domains used as full length IgG structures have constant domains derived from human IgG constant domains or specific variants thereof and are used as therapeutic molecules in a pharmaceutical preparation to prevent binding of OSM to cells displaying OSM receptor components.
  • the binding domains are configured as antibody fragments for use as a therapeutic molecule capable of prevent binding of OSM to cells displaying OSM receptor components.
  • a pharmaceutically acceptable formulation, delivery system, or kit or a method of treating oncostatin M-related conditions comprising one or more of the OSM binding domains of the invention such as but not limited to 13-28 and 30-46 and variants as provided by SEQ ID NO: 29 and 47.
  • FIG. 2A is a graph of the dose-response of the CHO cell-derived recombinant human and cynomolgous monkey oncostatin M in suppressing proliferation of A375-S2 cells as measured by BrdU incorporation and normalized to the control in the presence of vehicle only.
  • FIG. 2B is a graph showing the ability of the antibody M71, comprised of the L180 (SEQ ID NO: 53) and H17 (SEQ ID NO: 54) variable domains to relieve OSM suppression of A375-S2 proliferation where OSM was present at a concentration of 2 ng/ml.
  • FIG. 3 is a column graph showing the effect of M64, M71, M55, and M69 at 20 ⁇ g/ml in increasing 35 SO 4 -uptake, a measure of increased proteoglycan synthesis, above the level observed in the absence of antibody and where the non-specific isotype antibody was a control in co-cultures of human chondrocytes in alginate beads and human macrophages capable of secreting OSM.
  • FIG. 4A is a graph in showing the dose response of human OSM stimulated pSTAT3 in NHLF cells where the EC50 was found to be approximately 1 ng/ml.
  • FIG. 4B is a graph showing the ability of the antibody M71 to neutralize the pSTAT3 signal in the presence of 2 ng/ml OSM.
  • FIGS. 5A and B are scatter plots showing the amount of IP-10 (A) and MCP-1 (B) detected in the serum of individual mice after challenge with OSM and with or without pretreatment with the indicated concentration of M71 antibody.
  • FIGS. 6A and B are graphs showing the serum concentration over time in cynomolgous monkeys after intravenous (A) or subcutaneous (B) administration of 3 mg/Kg M71 or an Fc variant of M71.
  • BSA bovine serum albumin
  • CDR complementarity determining region
  • Cyno Cynomolgus monkey ( Macaca fascicularis );
  • DN diabetic nephropathy;
  • ECD extracellular domain;
  • IPF interstitial pulmonary arthritis;
  • L light chain;
  • Ig immunoglobulin;
  • Mab monoclonal antibody;
  • OSM oncostatin M;
  • OA osteoarthritis;
  • PBS phosphate buffered saline;
  • RA rheumatoid arthritis;
  • VL Variable light chain;
  • VH Variable heavy chain,
  • an “antibody” includes whole antibodies and any antigen binding fragment or a single chain thereof.
  • the antibody includes any protein or peptide containing molecule that comprises at least a portion of an immunoglobulin molecule, such as but not limited to, at least one complementarity determining region (CDR) of a heavy or light chain or a ligand binding portion thereof, a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework (FR) region, or any portion thereof, or at least one portion of a binding protein, which can be incorporated into an antibody of the present invention.
  • CDR complementarity determining region
  • antibody is further intended to encompass antibodies, digestion fragments, specified portions and variants thereof, including antibody mimetics or comprising portions of antibodies that mimic the structure and/or function of an antibody or a specified fragment or portion thereof, including single chain and single domain antibodies and fragments thereof.
  • Functional fragments include antigen-binding fragments to a preselected target.
  • binding fragments encompassed within the term “antigen binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH, domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH, domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR).
  • a Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH, domains
  • a F(ab′)2 fragment a bivalent fragment comprising two Fab fragments linked by a dis
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (I 988) Science 242:423-426, and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • scFv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody.
  • antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • libraries of scFv constructs can be used to screen for antigen binding capability and then, using conventional techniques, spliced to other DNA encoding human germline gene sequences.
  • One example of such a library is the “HuCAL: Human Combinatorial Antibody Library” (Knappik, A. et al. J Mol Biol (2000) 296(1):57-86).
  • CDR refers to the complementarity determining region or hypervariable region amino acid residues of an antibody that participate in or are responsible for antigen-binding.
  • the hypervariable regions or CDRs of the human IgG subtype of antibody comprise amino acid residues from residues 24-34 (L-CDR1), 50-56 (L-CDR2) and 89-97 (L-CDR3) in the light chain variable domain and 31-35 (H-CDR1), 50-65 (H-CDR2) and 95-102 (H-CDR3) in the heavy chain variable domain as described by Kabat et al. (1991 Sequences of Proteins of Immunological Interest, 5th Ed.
  • the numbering system of Chothia and Lesk takes into account differences in the number of residues in a loop by showing the expansion at specified residues denoted by the small letter notations, e.g., 30a, 30b, 30c, etc. More recently, a universal numbering system has been developed and widely adopted, international ImMunoGeneTics information System® (IMGT) (LaFranc, et al. 2005. Nucl Acids Res. 33:D593-D597).
  • IMGT ImMunoGeneTics information System
  • the CDRs are referred to in terms of both the amino acid sequence and the location within the light or heavy chain by sequential numbering.
  • location of the CDRs within the structure of the immunoglobulin variable domain is conserved between species and present in structures called loops, by using numbering systems that align variable domain sequences according to structural features, CDR and framework residues and are readily identified. This information is used in grafting and replacement of CDR residues from immunoglobulins of one species into an acceptor framework from, typically, a human antibody.
  • the term “maturation” is applied to directed changes in an antibody variable region for the purpose of altering the properties of the polypeptide.
  • antibodies achieve high affinity and specificity by the progressive process of somatic mutation. This process can be imitated in vitro to permit parallel selection and targeted variation while maintaining the sequence integrity of each antibody chain such that they reflect the species, in the present case, a human, antibody, while enhancing affinity or a biophysical parameter such as solubility or resistance to oxidation.
  • the process of making directed changes or “maturation” is typically performed at the level of the coding sequence and can be achieved by creating sublibraries for selection of the enhanced property.
  • OSM oncostatin M polypeptide or polynucleotide comprising a coding sequence encoding the OSM polypeptide.
  • Human OSM is the product of the human osm gene (Gene 5008).
  • epitope means a protein determinant capable of specific binding to an antibody.
  • Epitopes usually consist of chemically active surface groupings of molecules, such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and nonconformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • K D refers to the dissociation constant, specifically, the antibody K D for a predetermined antigen, and is a measure of affinity of the antibody for a specific target.
  • High affinity antibodies have a K D of 10 ⁇ 8 M or less, more preferably 10 M or less and even more preferably 10 ⁇ 10 M or less, for a predetermined antigen.
  • the reciprocal of K D is K A , the association constant.
  • K dis or “k 2 ,” or “k d ” as used herein, is intended to refer to the dissociation rate of a particular antibody-antigen interaction.
  • K D is the ratio of the rate of dissociation (k 2 ), also called the “off-rate (k off )” to the rate of association rate (k 1 ) or “on-rate (k on ).”
  • K D equals k 2 /k 1 or k off /k on and is expressed as a molar concentration (M). It follows that the smaller the K D , the stronger the binding. Thus, a K D of 10 ⁇ 6 M (or 1 microM) indicates weak binding compared to 10 ⁇ 9 M (or 1 nM).
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • the term also includes “recombinant antibody” and “recombinant monoclonal antibody” as all antibodies are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal or a hybridoma prepared by the fusion of antibody secreting animal cells and an fusion partner, (b) antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human or other species antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
  • an “isolated antibody,” as used herein, is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities.
  • An isolated antibody that specifically binds to an epitope, isoform or variant of human OSM may, however, have cross-reactivity to other related antigens, e.g., from other species (e.g., OSM species homologs).
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • a combination of “isolated” monoclonal antibodies having different specificities are combined in a well defined composition.
  • telomere binding refers to antibody binding to a predetermined antigen.
  • the antibody binds with a dissociation constant (K D ) of 10 ⁇ 7 M or less, and binds to the predetermined antigen with a K D that is at least twofold less than its K D for binding to a non-specific antigen (e.g., BSA, casein, or any other specified polypeptide) other than the predetermined antigen.
  • K D dissociation constant
  • an antibody recognizing an antigen and “an antibody specific for an antigen” are used interchangeably herein with the term “an antibody which binds specifically to an antigen.”
  • highly specific binding means that the relative K D of the antibody for the specific target epitope is at least 10-fold less than the K D for binding that antibody to other ligands.
  • type refers to the antibody class (e.g., IgA, IgE, IgM or IgG) that is encoded by heavy chain constant region genes.
  • Some antibody classes further encompass subclasses or “isotypes” which are also encoded by the heavy chain constant regions (e.g., IgG1, IgG2, IgG3 and IgG4).
  • Antibodies may befurther decorated by oligosaccharides linked to the protein at specific residues within the constant region domains which further enhance biological functions of the antibody. For example, in human antibody isotypes IgG1, IgG3 and to a lesser extent, IgG2, display effector functions as do murine IgG2a antibodies.
  • effector functions or “effector positive” is meant that the antibody comprises domains distinct from the antigen specific binding domains capable of interacting with receptors or other blood components such as complement, leading to, for example, the recruitment of macrophages and events leading to destruction of cells bound by the antigen binding domains of the antibody.
  • Antibodies have several effector functions mediated by binding of effector molecules. For example, binding of the C1 component of complement to antibodies activates the complement system. Activation of complement is important in the opsonisation and lysis of cell pathogens. The activation of complement stimulates the inflammatory response and may also be involved in autoimmune hypersensitivity.
  • Fc receptor Fc receptor
  • IgG gamma receptors
  • IgE eta receptors
  • IgA alpha receptors
  • IgM mi receptors
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • polypeptide means a molecule that comprises amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than 50 amino acids may be referred to as “peptides.” Polypeptides may also be referred as “proteins.”
  • the present invention provides isolated Mabs capable of binding and neutralizing the biological activity of OSM proteins and cleavage products.
  • the OSM binding mAbs of the invention are able to block OSM binding to gp130 or prevent the recruitment of LIFR ⁇ or OSMRb by OSM bound gp130.
  • the OSM mAb of the invention is capable of blocking OSM driven gp130 receptor signaling.
  • the human OSM gene product is a pre-pro-polypeptide 252 amino acids in length (SEQ ID NO: 11), having a signal peptide 25 amino acids in length and a proteolytic cleavage site between residues 234 and 235. It is a secreted protein having five cysteine residues forming two internal disulfides between residues 31 to 152 and 74 to 192 (Kallestad J C, et al. J Biol. Chem. 1991 May 15; 266 (14):8940-5). There are two potential N-linked glycosylation sites at residues 100 and 217, and when produced in eukaryotic cells, the protein is glycosylated. The human OSM has a free sulfhydryl at residue 105.
  • the predicted translation of the cloned sequence was found to be 99.6% identical to the predicted Macaca mullata (Rhesus) sequence, 92% identical to the human OSM protein sequence, and 41% identical to the mouse OSM protein sequence as disclosed in applicants copending application (U.S. Ser. No. 12/648,430).
  • the present invention is directed toward the identification of human derived OSM-binding Mabs capable of inhibiting downstream biologic activity resulting from OSM bound gp130 signaling and wherein the Mabs exhibit the ability to;
  • An OSM-neutralizing antibody of the invention is an antibody that inhibits, blocks, or interferes with at least one OSM activity or OSM receptor binding, in vitro, in situ and/or in vivo and does not promote, stimulate, induce, or agonize OSM activity or ligand binding nor does antibody binding mimic the downstream effects of OSM-driven ligation of OSM receptors, in particular gp130 interaction with OSM, such as signal transduction in a host cell.
  • a suitable OSM-neutralizing antibody, specified portion, or variant can also, optionally, affect at least one OSM activity or function, such as but not limited to; RNA, DNA or protein synthesis; protein release; cell activation, proliferation or differentiation; antibody secretion; OSM receptor signaling; OSM cleavage; OSM binding, OSM or gp130 induction, synthesis or secretion.
  • OSM activity or function such as but not limited to; RNA, DNA or protein synthesis; protein release; cell activation, proliferation or differentiation; antibody secretion; OSM receptor signaling; OSM cleavage; OSM binding, OSM or gp130 induction, synthesis or secretion.
  • the present invention is based upon the discovery of anti-human OSM monoclonal antibodies capable of inhibiting gp130 signaling after OSM binding or LIFR recruitment by OSM.
  • Antibody binding domains in the form of a Fab library displayed on filamentous phage particles linked to the pIX coat protein were selected for the ability to bind OSM.
  • a competition assay using gp130 was used to distinguish those Fabs that, when bound to OSM, prevented OSM from binding gp130.
  • Fabs were able to prevent LIFR recruitment to gp130 binding when bound to OSM.
  • a cell-based (A375, human melanoma cell) assay was used to identify several candidate antibodies capable of inhibiting gp130-mediated pSTAT3 activation of OSM expressing host cells.
  • OSM-binding antibodies described herein recognize at least two distinct regions on the active form of human OSM protein, indicating the additional discovery of multiple sites on OSM suitable for the targeting of antibodies or other compounds with similar function blocking capabilities.
  • expression and purification of the antibody binding domains provided herein as amino acid sequences further provides a tool which can provide the means for selection of novel molecules exhibiting OSM-neutralizing activity.
  • the anti-human OSM antibody has a binding region comprising a light chain variable (VL) or heavy chain variable (VH) region comprising the amino acid sequence as shown in SEQ ID NO: 49-55, and which antibody or binding portion thereof immunospecifically binds OSM.
  • VL light chain variable
  • VH heavy chain variable
  • the structural features of the antibodies exhibiting some or all of the above referenced biological activity as described herein and, in particular, the Mabs designated as M55 and M71 binding domains are used to create structurally related human anti-OSM antibodies that retain at least one functional property of the antibodies of the invention, such as binding to OSM. More specifically, one or more CDR regions of M55 and M71 (such as specified residues of SEQ ID NO: 1 and 8) can be combined recombinantly with known human framework regions and CDRs such as SEQ ID NO: 13-28, 30-46 to create additional, recombinantly-engineered, human anti-OSM antibodies of the invention.
  • the antibodies of the invention have the sequences, including FR1, 2, and/or 3; of IGVH1-69 (SEQ ID NO: 1) or of IGVH5-51 (SEQ ID NO: 3), wherein one or more residues from CDRs selected from the group consisting of SEQ ID NO: 13-22 are present in the CDR position of SEQ ID NO: 1 or 3, while still retaining the ability of the antibody to bind OSM (e.g., conservative substitutions).
  • the engineered antibody may be composed of one or more CDRs that are, for example, 90%, 95%, 98% or 99.5% identical to the CDRs listed in SEQ ID NOs: 13-22 or the variants of L-CDR3 as given by SEQ ID NO: 29 or 47.
  • engineered antibodies such as those described above, may be selected for their retention of other functional properties of antibodies of the invention, such as the ability to inhibit binding of OSM protein or a cleavage product thereof to GP130 positive cells to, which binding would result in suppression of proliferation of the GP130 positive cells in vivo.
  • Human monoclonal antibodies of the invention can be tested for binding to OSM by, for example, standard ELISA.
  • the epitope bound by the antibodies of the invention comprising as few as five to all of residues 51-227 of SEQ ID NO: 11 or a nucleic acid coding sequence therefore, can be used to immunize a subject in order to produce the antibodies of the invention directly in the host for the purpose of treating, preventing, or ameliorating disease or symptoms of disease associated with the production of OSM.
  • the human antibody is selected from a phage library, where that phage comprises human immunoglobulin genes and the library expresses human antibody binding domains as, for example, single chain antibodies (scFv), as Fabs, or some other construct exhibiting paired or unpaired antibody variable regions (Vaughan et lo al. Nature Biotechnology 14:309-314 (1996): Sheets et al. PITAS (USA) 95:6157-6162 (1998)); Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al. J. Mol. Biol., 222:581 (1991)).
  • scFv single chain antibodies
  • Human monoclonal antibodies of the invention can also be prepared using phage display methods for screening libraries of human immunoglobulin genes. Such phage display methods for isolating human antibodies are established in the art. See for example: U.S. Pat. Nos. 5,223,409; 5,403,484; and 5,571,698 to Ladner et al.; U.S. Pat. Nos. 5,427,908 and 5, 580,717 to Dower et al.; U.S. Pat. Nos. 5,969,108 and 6,172,197 to McCafferty et al.; and U.S. Pat. Nos. 5,885,793; 6,521,404; 6,544,731; 6,555,313; 6,582,915 and 6,593,081 to Griffiths et al.
  • Phage clones are selected by and identified through a multi-step procedure known as biopanning. Biopanning is carried out by incubating phage displaying protein ligand variants (a phage display library) with a target, removing unbound phage by a washing technique, and specifically eluting the bound phage. The eluted phage are optionally amplified before being taken through additional cycles of binding and optional amplification that enriches the pool of specific sequences in favor of those phage clones bearing antibody fragments that display the best binding to the target. After several rounds, individual phage clones are characterized, and the sequences of the peptides displayed by the clones are determined by sequencing the corresponding DNA of the phage virion.
  • biopanning is carried out by incubating phage displaying protein ligand variants (a phage display library) with a target, removing unbound phage by a washing technique, and specifically eluting the bound phage. The
  • a synthetic Fab library displayed on the pIX phage coat protein is used to select binder from a repertoire of human IgG sequences derived from human germline genes. Libraries were constructed on four VL and three VH domains encoded by known IGV and IGJ germline sequences selected based on the frequency which the sequences have been observed to be present in human antibodies isolated from natural sources.
  • VH The VH, IMGT nomenclature, selected are IGHV1-69 (SEQ ID NO: 1), IGHV3-23 (SEQ ID NO: 2), or IGHV5-51 (SEQ ID NO: 3).
  • the diversity in the VH design produces heavy chains with variable length sequence in the CDR3 region with limited diversity positions in the H-CDR1 and H-CDR2 which remain at a constant length.
  • Framework four (H-FR4) is held constant among all members of the library (SEQ ID NO: 4).
  • X 1 is A or G
  • X 2 is G or W
  • X 3 may be I or S
  • X 4 may be P or A
  • X 5 may be I or Y
  • X 6 may be F or N.
  • X 1 may be S, D, N, or T;
  • X 2 may be A, G, or W;
  • X 3 may be S or H;
  • X 4 may by V, A, N or G;
  • X 5 may be S, N, K or W;
  • X 6 may be Y, S, G, or Q;
  • X 7 may be S or D; and
  • X 8 may be S or G.
  • the H-CDR1 and H-CDR2 positions that were targeted for diversification were determined by 1) diversity in germline genes; and 2) frequency found in contact with antigen in antibody-antigen complexes of known structure (Almagro J Mol. Recognit. 17:132-143, 2004).
  • the amino acid diversity at the selected positions was determined by 1) usage in germline; 2) amino acids that are most frequently observed in human rearranged V genes; 3) amino acids predicted to be result from single base somatic mutations; and 4) biochemical and biophysical properties of amino acids that contribute to antigen recognition.
  • the library incorporates diversity in the CDR3 of the VH(H3) mimicking the repertoire of human antibodies (Shi et al. 2010 supra) as shown below (FORMULA I) where the final length is between 7 and 14 residues.
  • CDR3 amino acids glycine (G) and alanine (A) are frequently used in all positions.
  • aspartic acid (D) is frequently used in position 95 and tyrosine (Y) is frequently encoded in the positions preceding the canonical region of the J segment.
  • Amino acids phenylalanine (F), aspartic acid (D) and tyrosine (Y) predominate at positions 99-101 used in IgGs at these positions.
  • amino acids phenylalanine plus leucine (50/50 ratio) at position 99, aspartic acid at position 100 and tyrosine at position 101 were fixed.
  • sequence of Formula I is inserted between SEQ ID NOS: 1, 2, or 3 and SEQ ID NO: 4 to create a complete VH.
  • (D) Asp (D) and Gly (G) rich position.
  • (F) The Phe (F) dominant position.
  • the four light-chain library VL kappa genes are A27 (IGKV3-20*01), B3 (IGKV4-1*01), L6 (IGKV3-11*01), and 012 (IGKV1-39*01) where the gene name in parentheses are the presumed corresponding IMGT gene.
  • the Fabs are displayed on pIX via expression of a dicistronic vector wherein the VH-CH1 domain is fused to the coat protein sequence and the VL-CLkappa or VL-CLlambda is expressed as a free polypeptide which self-associates with the VH-CH1.
  • the CDR regions are underlined.
  • the VL CDR3 in all of the libraries has seven residues wherein the first two residues are glutamine (Gln, Q) and the residue corresponding to Kabat residue 95 is proline (Pro, P).
  • the sequence corresponds to QQX 1 X 2 X 3 X 4 PX 5 T (SEQ ID NO: 9), where varigation are as in the table below and at the residue positions are according to Kabat.
  • variable sequence varies in length from gene to gene
  • diversity in a particular residue location within a hypervariable loop or CDR can be described as follows using the residue numbering as defined in Al-Lazikani B, Lesk A M, Chothia C, 1997 (Standard conformations for the canonical structures of immunoglobulins. J Mol Biol 273: 927-948).
  • the changes in length of the hypervariable loops are accommodated by the designation of subpositions a, b, c, etc. for a given residue.
  • a framework 4 (FR4) segment such as JK4, FGQGTKVEIK (SEQ ID NO: 10) was used to form a complete human light chain variable region.
  • Methods for an integrated maturation process for improving binding parameters consisting of reshuffling VL or VH diversity or, alternatively, directed or limited VL modification are accomplished using the vectors and primers designed and used for the libraries as described in the referenced publication, as taught herein, and combined with what it known in the art.
  • OSM binding antibodies with the characteristics of the human Mabs disclosed herein may be made or binding fragments sourced from immunoglobulin domains formed by a number of methods, including the standard somatic cell hybridization technique (hybridoma method) of Kohler and Milstein (1975) Nature 256:495.
  • hybridoma method a mouse or other appropriate host animal, such as a hamster or macaque monkey, is immunized as described herein to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro.
  • Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • a suitable fusing agent such as polyethylene glycol
  • An OSM-neutralizing antibody can also be optionally generated by immunization of a transgenic animal (e.g., mouse, rat, hamster, non-human primate, and the like) capable of producing a repertoire of human antibodies, as described herein and/or as known in the art.
  • a transgenic animal e.g., mouse, rat, hamster, non-human primate, and the like
  • Cells that produce a human anti-OSM antibody can be isolated from such animals and immortalized using suitable methods, such as the methods described herein.
  • the antibody coding sequences may be cloned, introduced into a suitable vector, and used to transfect a host cell for expression and isolation of the antibody by methods taught herein and those known in the art.
  • transgenic mice carrying human immunoglobulin (Ig) loci in their germline configuration provide for the isolation of high affinity fully human monoclonal antibodies directed against a variety of targets including human self antigens for which the normal human immune system is tolerant (Lonberg, N. et al., U.S. Pat. No. 5,569,825, U.S. Pat. No. 6,300,129 and 1994, Nature 368:856-9; Green, L. et al., 1994, Nature Genet. 7:13-21; Green, L. & Jakobovits, 1998, Exp. Med. 188:483-95; Lonberg, N. and Huszar, D., 1995, Int. Rev. Immunol.
  • mice can be disrupted or deleted to eliminate the capacity of the animal to produce antibodies encoded by endogenous genes.
  • companies such as Abgenix, Inc. (Freemont, Calif.) and Medarex (San Jose, Calif.) can be engaged to provide human antibodies directed against a selected antigen using technology as described above.
  • Preparation of Polypeptides for Use as Target Ligands in Panning strategies and as immunogenic antigens can be performed using any suitable technique, such as recombinant protein production.
  • the target ligand or fragment thereof in the form of purified protein, or protein mixtures including whole cells or cell or tissue extracts, or, in the case of an immunization, the antigen can be formed de novo in the animal's body from nucleic acids encoding said antigen or a portion thereof.
  • the isolated nucleic acids of the present invention can be made using (a) recombinant methods, (b) synthetic techniques, (c) purification techniques, or combinations thereof, as well-known in the art.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using methods known in the art (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of human antibody regions). Where a hybridoma is produced, such cells can serve as a source of such DNA.
  • display techniques wherein the coding sequence and the translation product are linked such as phage or ribosomal display libraries, the selection of the binder and the nucleic acid is simplified.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria.
  • the invention further provides human immunoglobulins (or antibodies) which bind human OSM. These antibodies can also be characterized as engineered or adapted.
  • the immunoglobulins have variable region(s) substantially from a human germline immunoglobulin and include directed variations in residues known to participate in antigen recognition, e.g. the CDRs of Kabat or the hypervariable loops as structurally defined.
  • the constant region(s), if present, are also substantially from a human immunoglobulin.
  • the human antibodies exhibit K D for OSM of at least about 10 ⁇ 6 M (1 microM), about 10 ⁇ 7 M (100 nM), 10 ⁇ 9 M (1 nM), or less.
  • substitutions in either the CDR residues or other residues may be made.
  • the source for production of human antibody which binds to OSM is preferably the sequences provide herein as the variable regions, frameworks and/or CDRs, noted as SEQ ID NO: 13-55 identified as capable of binding human OSM and cross-reacting with cynomolgous monkey OSM using a repertoire of human derived Fab displayed on filamentous phage particles.
  • the substitution of any of the CDRs into any human variable domain framework is most likely to result in retention of their correct spatial orientation if the human variable domain framework adopts the same or similar conformation to the parent variable framework from which the CDRs originated.
  • the heavy and light chain variable framework regions to be paired in the final Mab can be derived from the same or different human antibody sequences.
  • the human antibody sequences can be the sequences of naturally occurring human antibodies, be derived from human germline immunoglobulin sequences, or can be consensus sequences of several human antibody and/or germline sequences.
  • Suitable human antibody sequences are identified by computer comparisons of the amino acid sequences of the mouse variable regions with the sequences of known human antibodies. The comparison is performed separately for heavy and light chains but the principles are similar for each.
  • variants can be generated using alternate and known methods for randomizing or variegating a nucleic acid sequence encoding the targeted residues within the variable domain.
  • Another method of determining whether further substitutions are required, and the selection of amino acid residues for substitution, can be accomplished using computer modeling.
  • Computer hardware and software for producing three-dimensional images of immunoglobulin molecules are widely available.
  • molecular models are produced starting from solved structures for immunoglobulin chains or domains thereof.
  • the chains to be modeled are compared for amino acid sequence similarity with chains or domains of solved three dimensional structures, and the chains or domains showing the greatest sequence similarity is/are selected as starting points for construction of the molecular model.
  • the solved starting structures are modified to allow for differences between the actual amino acids in the immunoglobulin chains or domains being modeled, and those in the starting structure.
  • the modified structures are then assembled into a composite immunoglobulin.
  • the model is refined by energy minimization and by verifying that all atoms are within appropriate distances from one another and that bond lengths and angles are within chemically acceptable limits.
  • nucleic acid sequences will encode each immunoglobulin amino acid sequence.
  • the desired nucleic acid sequences can be produced by de nova solid-phase DNA synthesis or by PCR mutagenesis of an earlier prepared variant of the desired polynucleotide. All nucleic acids encoding the antibodies described in this application are expressly included in the invention.
  • variable segments of human antibodies produced as described herein are typically linked to at least a portion of a human immunoglobulin constant region.
  • the antibody will contain both light chain and heavy chain constant regions.
  • the heavy chain constant region usually includes CH1, hinge, CH2, CH3, and, sometimes, CH4 domains.
  • the human antibodies may comprise any type of constant domains from any class of antibody, including IgM, IgG, IgD, IgA and IgE, and any subclass (isotype), including IgG1, IgG2, IgG3 and IgG4.
  • the constant domain is usually a complement-fixing constant domain and the class is typically IgG 1 .
  • the constant domain may be of the IgG 2 class.
  • the humanized antibody may comprise sequences from more than one class or isotype.
  • Nucleic acids encoding humanized light and heavy chain variable regions, optionally linked to constant regions, are inserted into expression vectors.
  • the light and heavy chains can be cloned in the same or different expression vectors.
  • the DNA segments encoding immunoglobulin chains are operably linked to control sequences in the expression vector(s) that ensure the expression of immunoglobulin polypeptides.
  • control sequences include a signal sequence, a promoter, an enhancer, and a transcription termination sequence (see Queen et al., Proc. Natl. Acad. Sci. USA 86, 10029 (1989); WO 90/07861; Co et al., J. Immunol. 148, 1149 (1992), which are incorporated herein by reference in their entirety for all purposes).
  • the present invention demonstrates that isolated monoclonal antibodies having the variable domains of M5, M6, M9, M10, M42, M45, M53, M54, M55, M62, M63, M65, M66, M67, M68, M69, M71, and M83 bind overlapping epitopes on OSM and display in vitro and/or in vivo OSM inhibiting activities.
  • the reactivity of the selected MAbs includes the ability to dose-dependently block OSM interaction with gp130, reduce OSM signaling in the presence of gp130, reduce OSM-stimulated proliferation of A375 cells, prevent macrophage-stimulated chondrocyte collagen production, or reduce cytokine release by OSM in vivo.
  • the antibodies or antigen binding fragments thereof are suitable both as therapeutic and prophylactic agents for treating or preventing OSM-associated conditions in humans and animals.
  • use will comprise administering a therapeutically or prophylactically effective amount of one or more monoclonal antibodies or antigen binding fragments of the present invention, or an antibody or molecule selected to have similar spectra of binding and biologic activity, to a susceptible subject or one exhibiting a condition in which OSM activity is known to have pathological sequelae, such as immunological disorders or tumor growth and metastasis.
  • Any active form of the antibody can be administered, including Fab and F(ab′)2 fragments.
  • the antibodies used are compatible with the recipient species such that the immune response to the MAbs does not result in an unacceptably short circulating half-life or induce an immune response to the MAbs in the subject.
  • the MAbs administered may exhibit some secondary functions, such as binding to Fc receptors of the subject and activation of ADCC mechanisms, in order to deplete the target cell population using cytolytic or cytotoxic mechanisms or they may be engineered to by limited or devoid of these secondary effector functions in order to preserve the target cell population.
  • Treatment of individuals may comprise the administration of a therapeutically effective amount of the antibodies of the present invention.
  • the antibodies can be provided in a kit as described below.
  • the antibodies can be used or administered as a mixture, for example, in equal amounts, or individually, provided in sequence, or administered all at once.
  • the dosage of administered agent will vary depending upon such factors as the patient's age, weight, height, sex, general medical condition, previous medical history, etc.
  • Another therapeutic use of the monoclonal antibodies of the present invention is the active immunization of a patient using an anti-idiotypic antibody raised against one of the present monoclonal antibodies.
  • Immunization with an anti-idiotype which mimics the structure of the epitope could elicit an active anti-OSM response (Linthicum, D. S. and Farid, N. R., Anti-idiotypes, Receptors, and Molecular Mimicry (1988), pp 1-5 and 285-300).
  • active immunization can be induced by administering one or more antigenic and/or immunogenic epitopes as a component of a vaccine.
  • Vaccination could be performed orally or parenterally in amounts sufficient to enable the recipient to generate protective antibodies against this biologically functional region, prophylactically or therapeutically.
  • the host can be actively immunized with the antigenic/immunogenic peptide in pure form, a fragment of the peptide, or a modified form of the peptide.
  • One or more amino acids, not corresponding to the original protein sequence can be added to the amino or carboxyl terminus of the original peptide, or truncated form of peptide.
  • Such extra amino acids are useful for coupling the peptide to another peptide, to a large carrier protein, or to a support
  • Amino acids that are useful for these purposes include: tyrosine, lysine, glutamic acid, aspartic acid, cysteine and derivatives thereof.
  • Alternative protein modification techniques may be used, e.g., NH2-acetylation or COOH-terminal amidation, to provide additional means for coupling or fusing the peptide to another protein or peptide molecule or to a support.
  • the antibodies capable of protecting against unwanted OSM bioactivity are intended to be provided to recipient subjects in an amount sufficient to effect a reduction, resolution, or amelioration in the OSM-related symptom or pathology.
  • An amount is said to be sufficient or a “therapeutically effective amount” to “effect” the reduction of symptoms if the dosage, route of administration, etc. of the agent are sufficient to influence such a response.
  • Responses to antibody administration can be measured by analysis of subject's affected tissues, organs, or cells as by imaging techniques or by ex vivo analysis of tissue samples.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient patient.
  • the OSM-neutralizing antibodies of the present invention can be used to measure or cause effects in an cell, tissue, organ or animal (including mammals and humans), to diagnose, monitor, modulate, treat, alleviate, help prevent the incidence of, or reduce the symptoms of, a condition mediated, affected or modulated by OSM or cells expressing OSM.
  • the present invention provides a method for modulating or treating at least one OSM related disease, in a cell, tissue, organ, animal, or patient, as known in the art or as described herein, using at least one OSM antibody of the present invention.
  • OSM is known to be up-regulated in a variety of disease states that involve inflammation and has been implicated in diverse biological roles including bone formation, cartilage degradation, cholesterol uptake, pain, and inflammation. Particular indications are discussed below.
  • OSM mediates cartilage destruction and shown that OSM causes chondrocyte degradation in intra-articular matrix from tissue explants. OSM also promotes cytokine release, such as TNF ⁇ , which is able to promote collagen release from cartilage as shown by T. Cawston et al (1998, Arthritis and Rheumatism, 41(10) 1760-1771) and that presently an antibody exemplified as M71 binding domains is capable of blocking systemic cytokine release.
  • An antibody of the present invention M55, M64, M69, and M71 demonstrated the ability to increase proteoglycan synthesis in a macrophage-chondrocyte co-culture system above the level seen in the absence of an OSM-specific antibody.
  • a neutralising anti-OSM antibody of the invention will inhibit OSM driven cytokine and chemokine release in vivo, such as IL-6, IP-10, and KC.
  • IP-10 interferon gamma-induced protein 10 kDa or small-inducible cytokine B10, is a protein that in humans is encoded by the CXCL10 gene (C-X-C motif chemokine 10 (CXCL10).
  • CXCL10 has been attributed to several roles, such as chemoattraction for monocytes/macrophages, T cells, NK cells, and dendritic cells, and promotion of T cell adhesion to endothelial cells.
  • CXCL1 chemokine (C-X-C motif) ligand 1
  • GRO1 oncogene GRO ⁇
  • NAP-3 Neutrophil-activating protein 3
  • MSGA- ⁇ melanoma growth stimulating activity, alpha
  • this protein is encoded by the CXCL1 gene.
  • CXCL1 is expressed by macrophages, neutrophils and epithelial cells, and has neutrophil chemoattractant activity.
  • an antibody or antibody fragment selected from the group M5, M6, M9, M10, M42, M45, M53, M54, M55, M62, M63, M65, M66, M67, M68, M69, M71, and M83 in the manufacture of a medicament for the treatment or prophylaxis of an articular proteoglycan degradative disease such as osteoarthritis, an inflammatory arthropathy or inflammatory disorder.
  • a particular use of an antagonist of OSM is in the manufacture of a medicament to prevent or reduce collagen release from cartilage.
  • the invention further provides a method for the treatment or prophylaxis of an inflammatory arthropathy or inflammatory disorder comprising administering an effective amount of such an antibody which blocks OSM binding to gp130 to a patient suffering from such a disorder.
  • An antibody of the invention may be used in a preparation for the treatment of pro-inflammatory process in which OSM directly or indirectly, such as through the release of inflammatory cytokines, leads to pathogenesis in tissues or organs especially in the skin, lungs, and joints.
  • pathologies include osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, neuropathic arthropathy, reactive arthritis, rotator cuff tear arthropathy, rheumatic fever, Reiter's syndrome, progressive systemic sclerosis, primary biliary cirrhosis, pemphigus, pemphigoid, necrotizing vasculitis, myasthenia gravis, multiple sclerosis, lupus erythematosus, polymyositis, sarcoidosis, granulomatosis, vasculitis, pernicious anemia, CNS inflammatory disorder, antigen-antibody complex mediated diseases, autoimmune hae
  • Those patients who are more particularly able to benefit from the method of the invention are those suffering from infection by E. coli, Haemophilus influenza B, Neisseria meningitides , staphylococci, or pneumococci.
  • Patients at risk for sepsis include those suffering from burns, wounds, renal or hepatic failure, trauma, burns, immunocompromised (HIV), hematopoietic neoplasias, multiple myeloma, Castleman's disease or cardiac myxoma.
  • fibrotic disease such as pulmonary fibrosis, diabetic nephropathy, idiopathic pulmonary fibrosis, systemic sclerosis, and cirrhosis.
  • Another indication for use of antibodies of the invention is in the treatment or prevention of nociceptive pain involving neurons of dorsal root ganglia.
  • the invention provides for stable formulations of an OSM-neutralizing antibody, which is preferably an aqueous phosphate buffered saline or mixed salt solution, as well as preserved solutions and formulations as well as multi-use preserved formulations suitable for pharmaceutical or veterinary use, comprising at least one OSM-neutralizing antibody in a pharmaceutically acceptable formulation.
  • Suitable vehicles and their formulation, inclusive of other human proteins, e.g., human serum albumin, are described, for example, in e.g. Remington: The Science and Practice of Pharmacy, 21st Edition, Troy, D. B. ed., Lipincott Williams and Wilkins, Philadelphia, Pa. 2006, Part 5, Pharmaceutical Manufacturing pp 691-1092, See especially pp. 958-989.
  • the OSM-neutralizing antibody in either the stable or preserved formulations or solutions described herein can be administered to a patient in accordance with the present invention via a variety of delivery methods including intravenous (I.V.); intramusclular (I.M.); subcutaneously (S.C.); transdermal; pulmonary; transmucosal; using a formulation in an implant, osmotic pump, cartridge, micropump; or other means appreciated by the skilled artisan, as well-known in the art.
  • I.V. intravenous
  • I.M. intramusclular
  • S.C. subcutaneously
  • transdermal pulmonary
  • transmucosal using a formulation in an implant, osmotic pump, cartridge, micropump; or other means appreciated by the skilled artisan, as well-known in the art.
  • site specific administration may be to body compartment or cavity such as intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, intralesional, vaginal, rectal, buccal, sublingual, intranasal, or transdermal means.
  • a dosage of antibody which is in the range of from about 1 ng/kg-100 ng/kg, 100 ng/kg-500 ng/kg, 500 ng/kg-1 ug/kg, 1 ug/kg-100 ug/kg, 100 ug/kg-500 ug/kg, 500 ug/kg-1 mg/kg, 1 mg/kg-50 mg/kg, 50 mg/kg-100 mg/kg, 100 mg/kg-500 mg/kg (body weight of recipient), although a lower or higher dosage may be administered.
  • an antagonist of the present invention will vary, depending upon factors such as the disease or disorder to be treated, the route of administration and the age and weight of the individual to be treated and the nature of the antagonist. Without being bound by any particular dosages, it is believed that for instance for parenteral administration, a daily dosage of from 0.01 to 20 mg/kg of an antibody (or other large molecule) of the present invention (usually present as part of a pharmaceutical composition as indicated above) may be suitable for treating a typical adult.
  • the treatment may be given in a single dose schedule, or preferably a multiple dose schedule in which a primary course of treatment may be with 1-10 separate doses, followed by other doses given at subsequent time intervals required to maintain and or reinforce the response, for example, at 1-4 months for a second dose, and if needed, a subsequent dose(s) after several months.
  • suitable treatment schedules include: (i) 0, 1 month and 6 months, (ii) 0, 7 days and 1 month, (iii) 0 and 1 month, (iv) 0 and 6 months, or other schedules sufficient to elicit the desired responses expected to reduce disease symptoms, or reduce severity of disease.
  • the antibodies of the present invention may be used alone or in combination with immunosuppressive agents such as steroids (prednisone etc.), cyclophosphamide, cyclosporin A or a purine analogue (e.g. methotrexate, 6-mercaptopurine, or the like), or antibodies such as an anti-lymphocyte antigen antibody, an anti-leukocyte antigen antibody, a TNF antagonist e.g. an anti-TNF antibody or TNF inhibitor e.g. soluble TNF receptor, or agents such as NSAIDs or other cytokine inhibitors.
  • immunosuppressive agents such as steroids (prednisone etc.), cyclophosphamide, cyclosporin A or a purine analogue (e.g. methotrexate, 6-mercaptopurine, or the like), or antibodies such as an anti-lymphocyte antigen antibody, an anti-leukocyte antigen antibody, a TNF antagonist e.g. an anti-TNF antibody or T
  • X 1 may be S, D, N, or T;
  • X 2 may be A, G, or W;
  • X 3 may be S or H;
  • X 4 may by V, A, N or G;
  • X 5 may be S, N, K or W;
  • X 6 may be Y, S, G, or Q;
  • X 7 may be S or D; and
  • X 8 may be S or G 3 Human IGHV5- EVQLVQSGAE VKKPGESLKI SCKGSGYSFT X 1 YWIX 2 WVRQM 51*01 PGKGLEWMG X 3 IX 4 PX 5 DSX 6 TRY SPSFQG QVTI
  • X 1 may be A, D, N, R or S
  • X 2 may be D, K, N, or S
  • X 3 may be A, D, F, H, N, S, W, or Y
  • X 4 may be A, D, K, G, F, T or N.
  • X 1 may be D, N, S, R or T
  • X 2 may be N, S, or R
  • X 3 may be A, D, H, N, S, or R
  • X 4 may be E, F, H, K, Q, S, or Y
  • X 5 may be A, D, G, or S.
  • X 1 may be A, F, H, S, or Y
  • X 2 may be E, K, N, or T
  • X 3 maybe A, D, F, H, N, S, W, or Y
  • X 4 may be A, D, S, W or Y.
  • X 1 may be A, D, F, H, P, S, or Y
  • X 2 may be D, E, G, H, I, K, N, R, T or Y
  • X 3 may be D, G, H, K, N, S, T, or R
  • X 4 may be F, I, L, N, R, W or Y as defined in Table 2.
  • Human OSM (NP — 065391 encoded by NM — 020530) is a 252 amino acid precursor processed into a full-length secreted protein of 227 amino acids (SEQ ID NO: 11), which is a proprotein further processed into the more fully active mature form, amino acids, 1-184.
  • Human OSM cDNA was ordered from OriGene (Cat. No.
  • Cynomolgous monkey OSM was cloned from cyno PBMC's RNA using Superscript III first strand synthesis system (InVitrogen) to obtain the cDNA and then PCR amplified using the UTR primers designed from the human OSM sequence as described in U.S. patent application Ser. No. 12/648,430.
  • the expressed full-length protein is shown in SEQ ID NO: 12 with the cleaved, active form represented by 184 residues, 51-227.
  • Precursor and mature forms of human and Cyno OSM were expressed in HEK 293 and purified using standard methodologies. The functional activities of the proteins were tested in the A375-S2 cell proliferation and pSTAT3 signaling assays using as control E. coli derived commercially available human OSM (R&D Systems, Cat. No. 295-OM).
  • a human IgG1 isotype antibody designated CNTO6234 was used.
  • Recombinant human OSM was biotinylated using NHS-ester chemistry (EZ-Link Sulfo-NHS-LC-Biotinylation Kit, Pierce, #21435) targeting amine residues on the cytokine.
  • the biotin-coupling reaction was optimized for a target labeling efficiency of one mole of biotin per mole of antigen. The latter minimizes the loss of binding and functional activity while ensuring near-complete labeling of the protein population.
  • the protein was purified from the free biotin reagent and residual leaving groups using Zeba Desalt Spin Columns included in the EZ-Link Sulfo-NHS-LC-Biotinylation Kit (Pierce). Approximately 80% of the starting material was recovered.
  • the HABA assay (Pierce Biotin Quantitation Kit, #28005) was used to measure the level of biotin incorporation, which indicated approximately one mole of biotin per mole of human OSM.
  • An Octet instrument (FortéBIO) was used to verify both streptavidin coupling and gp130-Fc (R&D Systems, Cat. No. 671-GP) binding for the biotinylated protein. Octet measurements showed that biotinylated human OSM bound gp130 with a profile essentially identical to that of the unlabeled starting material.
  • the 15 residue AviTag (GLNDIFEAQKIEWHE) (SEQ ID NO: 56) has similar biotin-acceptor kinetics as the endogenous BirA substrate BCCP (Beckett et. al. 1999, Protein Science). When tethered to a protein of interest the AviTag, having one acceptor lysine residue, will be biotinylated at only one position.
  • Recombinant Cyno OSM was biotinylated site-specifically in vitro using biotin-protein ligase and reagents commercially available from Avidity. The biotinylated Cyno OSM was purified using monovalent streptavidin affinity resin. The quality of the resulting protein was assessed by SDS-PAGE and SEC-HPLC.
  • the HABA assay (Pierce Biotin Quantitation Kit, #28005) was used to measure the level of biotin incorporation, which indicated approximately one mole of biotin per mole of cyno OSM.
  • An Octet instrument (FortéBIO) was used to verify both streptavidin coupling and gp130-Fc chimera binding for the biotinylated protein. Octet measurements showed that biotinylated Cyno OSM bound gp130 with a profile essentially identical to that of the unlabeled starting material.
  • Epitope binning is a competition assay performed in order to group the MAbs on the basis of binding characteristics performed using human IgG1 converted mAbs and the commercial antibody, MAB29, which is known to be an OSMR ⁇ /LIFR ⁇ recruitment-blocker (R-blocker).
  • the 384 multi-array plate was washed three times (PBS pH7.4, 0.05% Tween 20 (Scytek, PBT010) and to each well was added a 1.0 ⁇ g/ml solution of test mAb followed by shaking at level 6 on a titer plate shaker at room temperature for 1 hr. The plate was washed three times as before.
  • a 5 ⁇ g/ml solution of a competing mAb and 1 ⁇ g/ml biotinylated OSM were combined in a separate 96-well plate (COSTAR, 3357) in MSD assay buffer (1:3 of block buffer with PBS pH7.4, 0.05% Tween 20) and shaken at level 3 on a titer plate shaker at room temperature for 1 hr.
  • MSD assay buffer (1:3 of block buffer with PBS pH7.4, 0.05% Tween 20
  • the pre-complex of competing antibody and biotinylated OSM was added to each well of the 384 multi-array plate and shaken (level 6 on a titer plate shaker) at room temperature for 1 hr.
  • the plate was washed three times and to each well was added streptavidin sulfo TAG (MSD, R32Ad-S) and shaken (level 6 on a titer plate shaker) at room temperature for 30 min.
  • the plate was washed three times and to each well was added read buffer T diluted 1:4 with distilled water (MSD, R92TC-1).
  • the plate was read on an MSD 56000 instrument.
  • A375 cells are a human epithelia cell derived from malignant melanoma.
  • A375-S2 cells (ATCC; CRL-1872, a subline of CRL-1619 sensitive to IL-1) are cultured in complete growth medium (DMEM/GlutaMax-I, Gibco) supplemented with 10% FBS (Gibco) in T-175 culture flasks (Corning; Cat. No. 431080) and sub-cultured when approximately 80% confluent every three to four days in a 1:20 sub-culture ratio.
  • A375-S2 cells proliferation by human or Cynomolgus monkey OSM was measured by BrdU-incorporation using a chemiluminescent ELISA.
  • Oncostatin M reduces proliferation in the A375-S2 human melanoma cell line (Zarling et al. PNAS 83:9739-9743). This cell line was used to evaluate the anti-oncostatin M antibodies at all stages of antibody discovery.
  • A375-S2 cells were maintained in T-150 tissue culture flasks (BD Falcon 35-5001) in DMEM (Gibco 11995)+10% FBS (Gibco 16140)+1% Pen/Strep (Gibco 15140) at 37° C. in 95% O2—5% CO2 and split 1:10 twice per week.
  • cells were trypsinized (0.25%, Gibco 25200) to remove them from the T150 flasks and then plated at a density of 2000 cells/well in the inner 48 wells of black TC-coated plates (BD Falcon 353948) in 200 ⁇ L of DMEM/FBS/Pen-Strep. After overnight incubation at 37° C./95% O2—5% CO2, the media was removed and replaced with 180 ⁇ L of fresh media. A separate plate was prepared which contained all of the test solutions at 10 ⁇ the final concentrations. From this plate, 20 ⁇ L was transferred to the corresponding well in the cell plate to generate the appropriate experimental conditions. Each experimental condition was tested in triplicate.
  • the antibody and Oncostatin M were incubated together for at least 1 hour being added to cells.
  • the plates were then incubated at 37° C./95% O2—5% CO2 for an additional 72 hours.
  • the Chemiluminescent BrdU Cell Proliferation ELISA (Roche 11669915001) was performed.
  • the BrdU labeling reagent was added to the culture for 4 hours.
  • the media was then removed, and 100 ⁇ L of fix solution was added to each well. After 30 min at room temperature the solution was removed and 100 ⁇ L/well of anti-BrdU-POD solution was added. After 2 hours at room temperature, the plate was washed with PBS-Tween and 100 ⁇ L of Super Signal Pico (Thermo Scientific 37069) was added to each well. Luminescence was then read using a Perkin Elmer Victor3 instrument.
  • OSM inhibits the growth of A375-S2 cells by binding cell surface receptor gp130 and inducing receptor heterodimerization with OSMR ⁇ to initiate an intracellular signaling cascade that includes activation (phosphorylation) of the signaling molecule STAT3 (Kortylewski et al., Oncogene 18: 3742-3753, 1999). Disruption of STAT3 signaling abolishes OSM growth inhibition of A375-S2 cells, demonstrating that STAT3 activation is a key step in OSM signaling (Heinrich et al., Biochem. J. 374: 1-20, 2003). STAT3 phosphorylation in A375-S2 cells has been shown to be OSM concentration dependent and commercial kits are available to measure it in stimulated cells. Neutralization of OSM-induced STAT3 phosphorylation in A375-S2 cells was chosen as a primary screening assay for Mabs candidates.
  • A375-S2 cells are seeded into 96-well tissue culture plates (Corning; Cat. No. 3596) at 25,000 cells/well in 200 ⁇ l in complete growth media and incubated for 24 hours.
  • Cells are treated with a solution containing 5 ng/ml human OSM (recombinant, mammalian-cell derived, OSMN1-1) which has been pre-incubated for 3 hours at room temperature with 1:5 serially diluted experimental mAb starting at 10 ⁇ g/ml.
  • Controls include untreated cells, stimulated cells (5 ng/ml OSM only) and a hIgG1 isotype control mAb. All treatments are performed in triplicate unless otherwise noted.
  • the plate is sealed and placed on an orbital shaker (at 300 RPM) for 1 h at room temperature, washed three times with 150 ⁇ l of MSD wash buffer, and 25 ⁇ l of secondary detection antibody conjugate (anti-pSTAT3-Ru(bpy)32+) added to each well, again sealed and incubated on an orbital shaker (at 300 RPM) for 1 h at room temperature.
  • the plate is washed as previously and 150 ⁇ l of MSD read buffer (tripropylamine solution) is added to each well.
  • MSD read buffer tripropylamine solution
  • binding affinities were measured using Surface Plasmon Resonance (SPR) with a Biacore 3000 optical biosensor (Biacore) using human or Cyno OSM constructs as described.
  • a biosensor surface was prepared by coupling anti-IgG Fc antibody mixture of anti-Mouse (Jackson, Cat. No. 315-005-046) and anti-Human (Jackson, Cat. No. 109-005-098) to the carboxymethylated dextran surface of a CM-5 chip (Biacore, Cat. No. BR-1000-14) using the manufacturer's instructions for amine-coupling chemistry. Approximately 19,000 RU (response units) of anti-OSM antibody were immobilized in each of four flow cells.
  • the kinetic experiments were performed at 25° C. in running buffer (DPBS+0.005% P20+3 mM EDTA). Serial dilutions Human and Cyno OSM ECD from 100 nM to 0.412 nM were prepared in running buffer. About 200 RU of mAb were captured on flow cells 2 to 4 of the sensor chip. Flow cell 1 was used as reference surface. Capture of mAb was followed by a three-minute injection (association phase) of antigen at 50 ⁇ l/min, followed by 10 minutes of buffer flow (dissociation phase). The chip surface was regenerated by two pulses of 18-second injections of 100 mM H3PO4 (Sigma, Cat. No. 7961) at 50 ⁇ l/min.
  • the collected data was processed using the BIAevaluation software (Biacore, version 3.2). First, double reference subtraction of the data was performed by subtracting the curves generated by buffer injection from the reference-subtracted curves for analyte injections. Kinetic analysis of the data was performed using 1:1 binding model with global fit. The result for each mAb was reported in the format of Ka (On-rate), Kd (Off-rate) and K D (affinity constant).
  • the three heavy-chain library frameworks are combined with four light-chain library VL kappa frameworks: A27 (IGKV3-20*01 (SEQ ID NO: 5)), B3 (IGKV4-1*01 (SEQ ID NO: 6)), L6 (IGKV3-11*01 (SEQ ID NO: 7)), and 012 (IGKV1-39*01 (SEQ ID NO: 8)).
  • the Fabs V-regions are completed by the addition of a J-region (FR4) comprising SEQ ID NO: 4 in the heavy chains and SEQ ID NO: 10 in the light chains.
  • the heavy chain CDR3 is of variable length from 7-14 residues. Examples of the complete V-regions for each library are shown in FIG. 1 and numbered and CDR regions shown according to Kabat.
  • the initial set of anti-OSM phage display hits were identified using commercially available aglycosylated human OSM.
  • the Fab-pIX phage display libraries were panned using biotinylated human OSM (R&D Systems, Cat. No. 295-OM) capture on paramagnetic Streptavidin (SA) beads (Invitrogen, Cat. No. 112.05D) following a published protocol for phage selection (Marks and Bradbury, Antibody Engineering, Vol. 248: 161-176, Humana Press, 2004).
  • biotinylated human OSM was added to the phage libraries that had been pre-absorbed on unconjugated beads, to a final concentration of 100 nM and incubated for 1 hour with gentle rotation.
  • Blocked SA beads were added and incubated for 15 minutes to capture biotinylated OSM with bound phage.
  • the magnetically-captured phage/antigen/bead complex was washed 5 times with 1 ml of TBST and once with 1 ml TBS. Following removal of the final TBS wash, 1 ml of exponentially growing TG1 cells (Stratagene, Cat. No. 200123) was added and incubated at 37° C. for 30 minutes without shaking.
  • Infected bacteria were spread on LB/Agar (1% Glucose/100 mg/ml Carbenicillin) plates (Teknova, Cat. No. L5804) and incubated overnight at 37° C. Bacterial lawns were scraped and glycerol stocks prepared [15% glycerol/Carbenicillin (100 mg/ml)/2 ⁇ YT] and stored at ⁇ 80° C. To prepare phage for second-round panning, 25 ml of 2 ⁇ YT/Carbenicillin (100 ⁇ g/ml) was inoculated with 25 ⁇ l of bacterial glycerol stock and grown at 37° C. until an OD600 of roughly 0.5. Helper phage VCSM13 (Stratagene, Cat.
  • the panning parameters were: Round 1, 100 nM antigen, 1 hour incubation at room temperature, 5 ⁇ washes with TBST followed by 1 ⁇ wash with TBS; Round 2, 10 nM antigen, 1 hour incubation at room temperature, 10 ⁇ washes with TBST/1 ⁇ wash with TBS; and Round 3, 1 nM antigen, 16 hour (overnight) incubation at 4° C., 10 ⁇ washes with TBST/1 ⁇ wash with TBS.
  • the four-helix bundle architecture OSM is characterized by four ⁇ helical segments designated A, B, C and D linked by relatively unstructured loops.
  • OSM interacts with gp130 via an surface located in helices A and C (Site II) which was determined to include contact by amino acid residues Q16, Q20, G120, N123, N124 of SEQ ID NO: 1 (Deller et al. Structure 8(8): 863-874, 2000; Liu et al. Int. J. Mol. Med. 23: 161-172, 2009).
  • the surface responsible for OSM interaction with OSMR ⁇ and LIFR ⁇ Site III is believed to be largely defined by residues located in helix D (Deller et al. ibid).
  • the target affinity for a therapeutic lead was dictated by the need to compete decisively the OSM-gp130 interaction whose affinity (K D ) has been measured to be approximately 1 nM. Therefore, a target affinity for OSM of 100 pM or lower K D was desired.
  • Neutralizing MAbs M5, M6 and M9, found to be OSM-gp130 interaction blockers, plus M10 were chosen for inline maturation in order to produce derivates that met the 100 pM affinity requirement for a therapeutic candidate. It was an additional desired property that mAbs bind cynomolgus monkey OSM with an affinity within fivefold that for human OSM (K D of 500 pM or less).
  • compositions of the binding domains for these four Mabs were found to represent four unique heavy and light chain pairs designated as shown in Table 4.
  • each V-region is comprised of the designated CDR1 and CDR2 within the scaffold designated as SEQ ID NO: 1-3 or 5-8; followed by CDR3 (Table 3 and 4) and for the light chain variable region, respectively (Table 5 for LC and Table 6 for the HC) followed by a J-region as SEQ ID NO: 4 for the heavy chain and SEQ ID NO: 10 for the light chain.
  • Each of the four HC were comprised of a unique CDR3 (SEQ ID NOS: 19-22).
  • the LC variable regions of the selected Fabs all derive from the B3 library and include the curated germline sequence represented in the IMGT database as IGKV4-1 (B3) which was used as a starting sequence for the library diversification as described herein and referenced publications. Three of the four light chains had differences in CDR1 and had identical H-CDR2.
  • the L-CDR3 can be represented by a consensus sequence (SEQ ID NO: 29) represented by the formula Q-Q-(S,Y)-(F,Y)-S-(F,T)-PLT.
  • V-region pairings OSMM5, OSMM6, OSMM9 and OSMM10, described in Example 3 were chosen for light chain reselection in order to improve affinity.
  • an “in-line” maturation process described in Shi et al. J Mol Biol 397:385-396, 2010 and WO09085462A1 and U.S. Ser. No. 12/546,850 was used.
  • the V H regions of antigen-specific clones obtained in initial rounds of selection were combined with libraries of the corresponding V L scaffold, in this case the B3 based V-region (SEQ ID NO: 8).
  • V L library used in the primary selections as source of diverse V L chains
  • two additional libraries designed based on a recent analysis of the antigen-antibody complexes of known structure (Raghuanthan et al., J. Mol. Recognit, 2010). Residues were selected for diversification based on those residues most likely to be involved in the binding of the target protein also called specificity determining residue usage (SDRU). In the Vkappa light chains this has been determined to be three regions of contacts centered around the hypevariable loops as defined by Chothia and Lesk which differ slightly depending upon whether the target is a protein, a peptide, or a small hapten.
  • SDRU specificity determining residue usage
  • Table 5 shows the V L libraries used for affinity maturation, where B3 is the same library used during the discovery phase, Library 2 “SDRM focused” is based on the SDRU residues and focused diversity, and NNK is a randomized library.
  • “X” means any amino acid plus one stop codon generated by a NNK mix.
  • Table 7 summarizes the diversity in Libraries 1, 2 and 3.
  • amino acids were identified that were not part of the original design and thus were introduced as a consequence of the synthesis method.
  • these amino acids were: S (position 30c), T (position 30d), EK (position 30f, IW (position 32), TV (position 50), I (position 92), D (position 93), and F (position 96).
  • Fab-His protein was prepared from third-round panning outputs and monoclonal Fab-His ELISAs employed to identify individual hits with higher binding signal than the corresponding parental Fab.
  • Two concentrations of biotinylated human antigen were used in these ranking ELISAs: 2 nM and 0.2 nM.
  • the binding signal for each parental clone was set as 100%.
  • the VH of H14 and H17 share a common FR1-CDR1-FR2-CDR2-FR3 which is given by SEQ ID NO: 48 and comprising SEQ ID NO: 14 (CDR1) and SEQ ID NO: 17 (CDR2).
  • the biosensor surface was prepared by coupling each test mAb to the carboxymethylated dextran surface of a CM-5 chip (Biacore, Cat# BR-1000-14) following the manufacturer's instructions. Approximately 4,000-15,000 RU (response units) of each test mAb were immobilized in one the instrument's four flow cells. Competition experiments were performed at 25° C. in running buffer (DPBS+0.005% P20+3 mM EDTA). Human OSM (in-house, OSMN1-1) was diluted to 30 nM in running buffer and injected at 3 ⁇ l/min for 3 minutes over each of the flow cells with immobilized mAb.
  • association phase Following human OSM capture there was a three-minute injection (association phase) of either a competing mAb or human gp130-Fc at 300 nM followed by buffer flow (dissociation phase) for 3 minutes.
  • the chip surface was regenerated by two 12-second pulses of 100 mM H 3 PO 4 (Sigma, Cat#7961) at 50 ⁇ l/min.
  • the collected data were processed using BIAevaluation software version 3.2 (Biacore). First, the sensorgrams were aligned upon injection of human OSM. Then the binding levels (RU) for human OSM, competing mAb or gp130-Fc were recorded.
  • Flow-cell (Fc1, etc.) immobilized mAbs appear along the horizontal sample rows of Table 10.
  • M2 was previously shown to compete with the commercial antibody MAB295 which is known not to compete with gp130 binding to OSM.
  • the results demonstrate of the competition binding assays showed that M54, M55, M64, M69 and M71 compete with human gp130-Fc for OSM antigen (Table 11).
  • M71 was evaluated in a macrophage-chondrocyte co-culture system. Differentiated macrophages are known to produce Oncostatin M (Hasegawa et al. Rheumatology 38:612-617, 1999). OSM can decrease the synthesis of the highly-sulfated proteoglycan aggrecan (GAG) that makes up a significant portion of the cartilage matrix.
  • GAG highly-sulfated proteoglycan aggrecan
  • the anti-human Oncostatin M antibodies were discovered using recombinant HEK-generated His-Avi tagged human and cynomolgous monkey Oncostatin M and the activity of these antibodies was evaluated using these molecules as well as bacterial-derived recombinant human Oncostatin M from R&D Systems (295-OM). None of these are identical to native endogenous human Oncostatin M, either because of the his-avi tag (HEK-generated Osm) or the lack of glycosylation (bacterial recombinant Osm).
  • Macrophages are known to secrete Oncostatin M (Grove et al., J Lipid Res 32:1889-97, 1991) and oncostatin M decreases proteoglycan synthesis in human chondrocytes (Sanchez et al. OA and Cart. 12:810-10, 2004).
  • a macrophage-chondrocyte co-culture system was used to determine the ability of the anti-human Oncostatin M antibodies to neutralize endogenous, or native, human Oncostatin M. Briefly, single alginate beads containing 40,000 normal human articular chondrocytes were cultured for 72 hours in the presence of differentiated human macrophages. These experiments were conducted in the presence of a dose-range of anti-Oncostatin M antibody. At the end of the experiment, proteoglycan synthesis was measured by the incorporation of radioactive 35 SO 4 .
  • CD14+ peripheral blood monocytes were obtained from AllCells.
  • the monocytes were cultured on 48-well plates at 2.5 ⁇ 105 cells/well in 0.5 mls of macrophage medium (RPMI+Glutamine with 10% heat inactivated FBS, 1% NEAA and 1% Pen-Strep).
  • the cells were treated with 100 ng/ml of macrophage-colony stimulating factor (M-CSF). After 48 hours the media was replaced to remove non-adherent cells. On day 6, the M-CSF containing macrophage medium was replaced with macrophage medium without M-CSF.
  • M-CSF macrophage-colony stimulating factor
  • the macrophage medium was replaced with chondrocyte medium (50% Ham's F-12/50% DMEM with 10% fetal calf serum) and a single alginate bead chondrocyte culture (Articular Engineering #CDD-H-2200) was added to each well.
  • the aliquots of conditioned macrophage medium were reserved and stored at ⁇ 80° C. for analysis of Oncostatin M levels using the R&D Systems Human Oncostatin M DuoSet (DY295).
  • the alginate bead-macrophage co-cultures were maintained in the presence of either 20 ⁇ g/ml of anti-human Oncostatin M antibodies (M64, M71, M55, M69) or in the presence of a dose-range (5 ⁇ g/ml to 0.00076 ⁇ g/ml; 1 to 3 dilutions) of antibody (M71 and M55).
  • human IgG1 isotype control (CNTO6234) was included on the plate at the highest concentration of anti-oncostatin M antibody tested.
  • Other controls were chondrocytes only (no co-culture) and chondrocytes in the presence of 2 ng/ml human Oncostatin M.
  • M64, M71, M55, and M69 increased proteoglycan synthesis above the level observed in the absence of antibody while the isotype control had no effect ( FIG. 3 ).
  • M71 dose-dependently increased proteoglycan synthesis to the level exhibited by chondrocytes alone (defined as 100% neutralization), with an EC50 of 30 ng/ml and the isotype control had no effect.
  • OSM induces proliferation and collagen production in normal human lung fibroblasts (Scaffidi et al., Br. J. Pharmacol 136: 793-801, 2002). Over-production of collagen by fibroblasts is a key feature of a number of pathological conditions (Lim et al. Oncogene 23(39): 5416-25, 2006; Huang et al. J Cell Biochem 81(1): 102-13, 2001).
  • Oncostatin M receptor signaling activates the JAK-STAT pathway and phosphorylation of STAT3 is an early event in the signaling pathway (Marche et al. (1997) Signaling of Type II Oncostatin M Receptor. J Biol Chem 272:15760-15764).
  • Oncostatin M was determined in normal human lung fibroblasts (NHLF) using the R&D Systems human/mouse pSTAT3 Duoset (DYC4607-5). This assay was then used to determine the ability of M55 and M71 to neutralize Oncostatin M signaling.
  • NHLF from Lonza (CC-2512) grown in Lonza proprietary FGM-2 (CC-3132) media. Briefly, the cells were plated at 25,000 cells/well in FGM-2 and cultured for 24 hours. The cells were then treated with Oncostatin M or antibody plus Oncostatin M for 10 minutes. In order to avoid temperature dependent effects during this 10 minute incubation, all of the solutions used to prepare the treatments were pre-warmed and maintained at 37° C. After the 10 minute treatment, media was aspirated away and replaced with complete lysis buffer.
  • lysis buffer was prepared by adding 1 tablet of Protease Inhibitor Cocktail (Roche, 11836153001) and 110 ⁇ L HALT phosphatase inhibitor (Thermo Scientific 78420) to 11 mls of lysis buffer. After the 10 minute lysis step, the resulting lysate was ready for pSTAT3 detection.
  • NHLF cells were treated with a dose-range of OSM (100 ng/ml to 0.024 ng/ml w/1 to 4 dilutions, triplicate wells).
  • a dilution plate was prepared in pre-warmed PBS+1% BSA in which the concentration of Osm in each well was 10 ⁇ higher than the final concentration needed for treatment, with media-only wells included as untreated controls.
  • the media was completely removed from the culture plate and replaced with 180 ⁇ l of pre-warmed FGM-2.
  • a timer was started for 10 minutes and then 20 ⁇ l was transferred from each well in the dilution plate to the corresponding well in the culture plate.
  • the treatment solutions in the cell plate were completely removed by aspiration and replaced with 100 ⁇ l of complete lysis buffer.
  • the lysis buffer was added to wells in the same order as the treatments.
  • the assay plate was then placed on a shaker for 10 minutes. After shaking, the lysates were either frozen at ⁇ 80° C. for later testing or transferred directly to an ELISA plate coated with anti-pSTAT3.
  • the ELISA (R&D Systems human/mouse pSTAT3 Duoset) was carried out according to the manufacturer's instructions with the exceptions that 1) only 90 ⁇ l of lysate or standard was added to each well and 2) SuperSignal Pico (ThermoScientific 37069) was used as the HRP substrate.
  • the ELISA plate was read for luminescence on a Victor3 plate reader.
  • the cells were treated with a dose-range of anti-OSM antibody (triplicate wells, 500 ng/ml to 0.005 ng/ml with 1-10 dilutions or 50 ng/ml to 1.563 ng/ml with 1 to 2 dilutions) in the presence of 2 ng/ml human Osm.
  • a dilution plate was prepared in PBS for the antibody dose-response treatment at 20 ⁇ the final concentration, with wells included for the isotype control (at the highest concentration of the anti-OSM antibody) as well as with no antibody for both the untreated control and the cells treated only with OSM.
  • Human oncostatin M was prepared separately at 40 ng/ml (20 ⁇ the final concentration) in FGM-2.
  • the 20 ⁇ antibody and OSM solutions were mixed in equal volumes in the dilution plate to generate 10 ⁇ treatment solutions and the plate was incubated for 1 hour at 37° C. to allow OSM to bind to the antibody. After 1 hour, the media was removed from the culture plate and replaced with 180 ⁇ l of pre-warmed FGM-2. A timer was started for 10 minutes and 20 ⁇ l was transferred from each well in the dilution plate to the corresponding well in the culture plate. After 10 minutes, the solutions in the cell plate were completely removed by aspiration and replaced with 100 ⁇ l of complete lysis buffer.
  • the lysis buffer was added to wells in the same order as the treatments.
  • the assay plate was then placed on a shaker for 10 minutes. After shaking, the lysates were either frozen at ⁇ 80° C. for later testing or transferred directly to an ELISA plate coated with anti-pSTAT3.
  • the ELISA (R&D Systems human/mouse pSTAT3 Duoset) was carried out according to the manufacturer's instructions with the exceptions that 1) only 90 ⁇ l of lysate or standard was added to each well and 2) SuperSignal Pico (ThermoScientific 37069) was used as the HRP substrate.
  • the ELISA plate was read for luminescence on a Victor3 plate reader.
  • Human Oncostatin M increased pSTAT3 in NHLF cells with an EC50 of approximately 1 ng/ml.
  • An example of an Oncostatin M dose response is provided in FIG. 4A .
  • the EC50 was 0.90 ng/ml with a 95% confidence interval from 0.70 to 1.17 ng/ml.
  • the ability of the anti-oncostatin M antibodies to neutralize was determined in the presence of 2 ng/ml of Oncostatin M and all of the data were normalized to the luminescence in the presence of 2 ng/ml OSM with no antibody present.
  • FIG. 4B shows the dose-dependent neutralization of OSM-induced STAT3 phosphorylation by M71.
  • the dose-response curve was calculated from data from six separate experiments and the calculated IC50 for M71 was 8.9 ng/ml with a 95% confidence interval from 6.9 to 11.6 ng/ml.
  • Oncostatin M71 was evaluated for its ability to block the production of cytokines in vivo after systemic administration human Oncostatin M. Intraperitoneal injection of human Oncostatin M increases levels of certain serum cytokines, presumably through and interaction with the murine leukemia inhibitory factor receptor.
  • mice Systemic (i.p.) administration of human oncostastin M to mice was developed as a model to evaluate the ability of the anti-oncostatin M monoclonal antibodies to neutralize in an in vivo setting.
  • Mice were injected i.p. with 10 ⁇ g of human Oncostatin M in 200 ⁇ l of PBS or PBS vehicle control. After 1 hour the mice were anesthetized with CO2 and blood was collected by terminal cardiac puncture. The individual blood samples were allowed to clot on ice for 20 min and then spun at 3500 rpm for 10-15 minutes. Serum samples were kept frozen until analyzed, as per the manufacturer's instructions, with the Milliplex Murine MAP Cytokine/Chemokine Multiplex (32) Panel.
  • mice were dosed subcutaneously with M71 or M55 (human IgG1 anti-human Osm at 20, 2.0 or 0.2 mg/kg), CNTO6234 (huIgG1 isotype control, 20 mg/kg) or PBS in a volume of 10 ⁇ l/g. After 24 hours, each mouse was then injected i.p. with either 10 ⁇ g of in-house generated CHO cell-derived recombinant human oncostatin M in PBS (Sigma D8357) w/0.1% mouse serum albumin (Sigma A3559) or with PBS-MSA vehicle control alone (200 ⁇ L total volume).
  • mice After 1 hour the mice were anesthetized with CO2 and blood was collected by terminal cardiac puncture. The individual blood samples were allowed to clot on ice for 20 min and then spun at 3500 rpm for 10-15 minutes. Serum samples were kept frozen until analyzed using the Milliplex Murine MAP Cytokine/Chemokine Multiplex (32) Panel. Serum samples were analyzed as per the manufacturer's instructions.
  • IP-10 interferon gamma-induced protein 10 kDa or small-inducible cytokine B10
  • CXCL10 C-X-C motif chemokine 10
  • CXCL10 has been attributed to several roles, such as chemoattraction for monocytes/macrophages, T cells, NK cells, and dendritic cells, and promotion of T cell adhesion to endothelial cells.
  • CXCL1 chemokine (C-X-C motif) ligand 1
  • GRO1 oncogene GRO ⁇
  • NAP-3 Neutrophil-activating protein 3
  • MSGA- ⁇ melanoma growth stimulating activity, alpha
  • this protein is encoded by the CXCL1 gene.
  • CXCL1 is expressed by macrophages, neutrophils and epithelial cells, and has neutrophil chemoattractant activity.
  • a Fab fragment comprising the V-regions H17 (SEQ ID NO: 51) and L180 (SEQ ID NO: 55) was crystallized with human OSM (SEQ ID NO: 10) residues 26-212.
  • OSM shares its four-helix bundle three-dimensional structure with other members of the gp130 cytokine family.
  • the four-helix bundle architecture is characterized by four ⁇ helical segments designated A (residues 10-37), B (residues 67-90), C (residues 105-131) and D (residues 159-185) linked by relatively unstructured loops.
  • OSM interacts with gp130 via an epitope termed Site II which includes amino acid residues (Q16, Q20, G120, N123, N124) located in helices A and C (Deller et al. Structure 8(8): 863-874, 2000; Liu et al. Int. J. Mol. Med.
  • Site III the epitope responsible for OSM interaction with OSMR ⁇ and LIFR ⁇ , is believed to be largely defined by residues located in helix D (Deller et al. Structure 8(8): 863-874, 2000).
  • Crystallization of the complex was performed by the sitting-drop vapor-diffusion method at 20° C. using the Oryx4 protein crystallization robot (Douglas Instruments), dispensing equal volumes of 0.2 ⁇ L protein complex (10.95 mg/mL) and 0.2 ⁇ L reservoir solution. Multiple crystallization screens were performed. The majority of droplets remained clear, reflecting the high solubility of the complex. Crystals were obtained from 0.1 M MES pH 6.5, 2.4 M ammonium sulfate and 0.1 M Tris pH 8.5, 3.5 M sodium formate.
  • the OSM residues in contact with H14/L180 Fab constitute the binding epitope.
  • the antibody residues in contact with OSM constitute the binding paratope.
  • All six CDRs of the two variable domains are involved in OSM binding. Residues in contact are given in Table 13 and shown in FIG. 5 .
  • the long CDR-L1 together with the CDRs of the heavy chain variable region H1, form a valley-like antigen binding site with a small ridge at the bottom ( FIG. 4C , left panel).
  • the two sides of the valley embrace the OSM four-helix bundle along helices A and C with the bottom ridge binding in between the two helices ( FIG. 4C , right panel).
  • the antibody and antigen binding interface buries 2,514 ⁇ 2 of solvent accessible surface (1225 ⁇ 2 for Ab and 1298 ⁇ 2 for Ag). Although there are a number of charges residues at the interface, there is no charge-charge pairing, suggesting vdw and H-bonding play the most important roles in antibody and antigen interactions.
  • OSM is a soluble target associated with the inflammatory processes as opposed to a cell-surface displayed target on a cell.
  • the nature of a complete IgG comprising the binding regions as discovered herein and additionally having an Fc domain can thus be tailored to the purpose and therapeutic specification related to its use using methods of engineering the Fc with mutations conferring altered FcR binding.
  • a sustained activity and persistence in the circulation is a beneficial specification of a therapeutic monoclonal IgG. Therefore, an Fc-domain having enhanced affinity for the neonatal receptor (FcRn).
  • M428L MedImmune U.S. Pat. No. 7,670,600
  • N434S U.S. Pat. No. 7,371,826, WO2006/053301
  • M71 and M71 L/S were compared in standard activity assays and compared for persistence in the circulation of a non-human primate.
  • M71 and M71 L/S were compared side-by-side in the A375-S2 proliferation assay. Dose-responses were evaluated from a starting concentration of 1 ⁇ g/ml, with 1 to 5 dilutions to 0.00032 ⁇ g/ml. At a concentration of 1 ⁇ g/ml the isotype controls for these antibodies, CNTO3930 and CNTO8852, respectively, had no effect on the ability of 2 ng/ml of human Oncostatin M to inhibit proliferation. Both M71 and M71 L/S completely neutralized the effect of Oncostatin M at 1 ⁇ g/ml, with no measurable difference in IC50.
  • mice were dosed subcutaneously with M71 and M71 L/S (20, or 5.0 mg/kg), CNTO3930 (huIgG1 isotype control, 20 mg/kg), CNTO8852 (isotype control for Fc mutated version, 20 mg/kg) or PBS in a volume of 10 ⁇ l/g. After 24 hours, each mouse was injected i.p. with either 10 ⁇ g of in-house generated CHO cell-derived recombinant human oncostatin M in PBS (Sigma D8357) w/0.1% mouse serum albumin (Sigma A3559) or with PBS-MSA vehicle control alone (200 ⁇ L total volume).
  • mice were anesthetized with CO2 and blood was collected by terminal cardiac puncture.
  • the individual blood samples were allowed to clot on ice for 20 min and then spun at 3500 rpm for 10-15 minutes. Serum samples were kept frozen until analyzed using a custom Millipore murine multiplex consisting of beads specific for IL-6, MCP-1, eotaxin, KC and IP-10.
  • Neither isotype control had any effect on cytokine release induced by human oncostatin M.
  • both M71 and M71 L/S neutralized oncostatin M-induced cytokine release, with no apparent differences in potency or efficacy.
  • the serum half-life of M71 and M71 L/S were compared in a non-terminal cynomolgous monkey pharmacokinetics study.
  • Antibody levels in the serum were determined using an ELISA optimized in cynomologous monkey serum for the MesoScale Discovery platform.
  • the biotinylated capture antibody was an anti-idotype antibody raised against M71 (mouse anti-M71).
  • the detection antibody was ruthenium-labeled anti-human IgG and the readout was MesoScale Discovery chemiluminescence.
  • FIGS. 6A and B The results of the study are shown in FIGS. 6A and B.
  • FIG. 6A shows the plots from the i.v. dosing where the serum half-life of M71 was 15.21+/ ⁇ 3.0 days and the half-life of M71 L/S was 29.4+/ ⁇ 2.3 days. Similar results were obtained from s.c. dosing ( FIG. 6B ) with a serum half-life of 15.4+/ ⁇ 4 days for M71 and 32.0+/ ⁇ 5.9 days for M71 L/S.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Diabetes (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Dermatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurology (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
US13/269,976 2010-10-13 2011-10-10 Human Oncostatin M Antibodies and Methods of Use Abandoned US20120093833A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/269,976 US20120093833A1 (en) 2010-10-13 2011-10-10 Human Oncostatin M Antibodies and Methods of Use
US14/104,520 US9163083B2 (en) 2010-10-13 2013-12-12 Human oncostatin M antibodies
US14/870,774 US9587018B2 (en) 2010-10-13 2015-09-30 Polynucleotides encoding human oncostatin M antibodies
US15/423,189 US10179812B2 (en) 2010-10-13 2017-02-02 Method of treating idiopathic pulmonary fibrosis by administering human oncostatin M antibodies
US16/214,891 US10941197B2 (en) 2010-10-13 2018-12-10 Method of treating osteoarthritis with human oncostatin M antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US39268310P 2010-10-13 2010-10-13
US13/269,976 US20120093833A1 (en) 2010-10-13 2011-10-10 Human Oncostatin M Antibodies and Methods of Use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/104,520 Continuation US9163083B2 (en) 2010-10-13 2013-12-12 Human oncostatin M antibodies

Publications (1)

Publication Number Publication Date
US20120093833A1 true US20120093833A1 (en) 2012-04-19

Family

ID=45934343

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/269,976 Abandoned US20120093833A1 (en) 2010-10-13 2011-10-10 Human Oncostatin M Antibodies and Methods of Use
US14/104,520 Active US9163083B2 (en) 2010-10-13 2013-12-12 Human oncostatin M antibodies
US14/870,774 Active US9587018B2 (en) 2010-10-13 2015-09-30 Polynucleotides encoding human oncostatin M antibodies
US15/423,189 Active US10179812B2 (en) 2010-10-13 2017-02-02 Method of treating idiopathic pulmonary fibrosis by administering human oncostatin M antibodies
US16/214,891 Active 2032-01-15 US10941197B2 (en) 2010-10-13 2018-12-10 Method of treating osteoarthritis with human oncostatin M antibodies

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/104,520 Active US9163083B2 (en) 2010-10-13 2013-12-12 Human oncostatin M antibodies
US14/870,774 Active US9587018B2 (en) 2010-10-13 2015-09-30 Polynucleotides encoding human oncostatin M antibodies
US15/423,189 Active US10179812B2 (en) 2010-10-13 2017-02-02 Method of treating idiopathic pulmonary fibrosis by administering human oncostatin M antibodies
US16/214,891 Active 2032-01-15 US10941197B2 (en) 2010-10-13 2018-12-10 Method of treating osteoarthritis with human oncostatin M antibodies

Country Status (20)

Country Link
US (5) US20120093833A1 (ja)
EP (1) EP2627356B1 (ja)
JP (3) JP6216917B2 (ja)
KR (1) KR101920250B1 (ja)
CN (1) CN103261223B (ja)
AR (2) AR083418A1 (ja)
AU (1) AU2011313847B2 (ja)
BR (1) BR112013009083B1 (ja)
CA (1) CA2814652C (ja)
EA (1) EA031044B1 (ja)
IL (1) IL225573B (ja)
JO (1) JO3496B1 (ja)
MX (1) MX354095B (ja)
RU (1) RU2600444C2 (ja)
SG (2) SG189322A1 (ja)
TW (1) TWI548749B (ja)
UA (1) UA118646C2 (ja)
UY (1) UY33670A (ja)
WO (1) WO2012051111A2 (ja)
ZA (1) ZA201303429B (ja)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013166011A2 (en) 2012-05-02 2013-11-07 Janssen Biotech, Inc. Binding proteins having tethered light chains
US9550828B2 (en) 2013-09-05 2017-01-24 Boise State University Oncostatin M (OSM) antagonists for preventing cancer metastasis and IL-6 related disorders
US20170327573A1 (en) * 2014-09-24 2017-11-16 Universitá Degli Studi Di Padova Composition to induce bone marrow stem cell mobilization
WO2020127884A1 (fr) * 2018-12-21 2020-06-25 Universite De Poitiers PROTEINE DE LIAISON SPECIFIQUE CAPABLE DE SE LIER SPECIFIQUEMENT A L'ONCOSTATINE M HUMAINE (hOSM) ET SES UTILISATIONS
US11633457B2 (en) 2019-04-11 2023-04-25 Boise State University Pharmaceutical compositions comprising oncostatin m (OSM) antagonist derivatives and methods of use

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120093833A1 (en) * 2010-10-13 2012-04-19 Juan Carlos Almagro Human Oncostatin M Antibodies and Methods of Use
PT2643352T (pt) * 2010-11-23 2018-08-01 Glaxo Group Ltd Proteínas de ligação a antigénio para a oncostatina m (osm)
CN104083754A (zh) * 2014-07-08 2014-10-08 武汉大学 Ⅱ型抑瘤素m受体(osmr)在脑卒中疾病中的功能和应用
CN104258396A (zh) * 2014-09-29 2015-01-07 武汉大学 Ⅱ型抑瘤素m受体在治疗动脉粥样硬化中的功能和应用
EP3250599B1 (en) 2015-01-29 2021-04-14 Oxford University Innovation Limited Biomarker
EP3390453A2 (en) * 2015-12-17 2018-10-24 Janssen Biotech, Inc. Antibodies specifically binding hla-dr and their uses
GB201614627D0 (en) * 2016-08-30 2016-10-12 Glaxosmithkline Ip Dev Ltd Antigen binding proteins
WO2019084307A1 (en) * 2017-10-26 2019-05-02 Celldex Therapeutics, Inc. ANTI-MERTK ANTIBODIES AND METHODS OF USE
CN109884320A (zh) * 2019-03-29 2019-06-14 重庆医科大学 抑瘤素m作为生物标志物在制备脓毒症诊断试剂中的应用
WO2020208816A1 (ja) 2019-04-12 2020-10-15 住友電気工業株式会社 光ファイバテープ心線、ダイス、および、光ファイバテープ心線の製造方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070286861A1 (en) * 2004-03-30 2007-12-13 Ellis Jonathan H Immunoglobulins
US20110092372A1 (en) * 2009-10-14 2011-04-21 Juan Carlos Almagro Methods of Affinity Maturing Antibodies

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5681930A (en) 1985-12-20 1997-10-28 Bristol-Myers Squibb Company Anti-oncostatin M monoclonal antibodies
US5618715A (en) 1985-12-20 1997-04-08 Oncogen Limited Partnership Oncostatin M and novel compositions having anti-neoplastic activity
US5451506A (en) 1985-12-20 1995-09-19 Oncogen Limited Partnership Oncostatin M and novel compositions having anti-neoplastic activity
US5120535A (en) 1986-11-26 1992-06-09 Oncogen Oncostatin M and novel compositions having anti-neoplastic activity
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5202116A (en) 1989-04-10 1993-04-13 Oncogen Methods for controlling human endothelial cell proliferation and effector functions using oncostatin m
ZA909842B (en) 1989-12-08 1991-09-25 Oncogen Proteins with oncostatin m activity and process for their preparation
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
IL97623A (en) 1990-03-29 1996-01-19 Bristol Myers Squibb Co Monoclonal antibodies against oncostatin M, their preparation and pharmaceutical preparations containing them
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
KR100272077B1 (ko) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 이종 항체를 생산할 수 있는 전이유전자를 가진 인간이외의 동물
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
GB9019553D0 (en) * 1990-09-07 1990-10-24 Unilever Plc Specific binding agents
PT1696031E (pt) 1991-12-02 2010-06-25 Medical Res Council Produção de auto-anticorpos a partir de reportórios de segmentos de anticorpo e exibidos em fagos
US5585793A (en) 1994-06-10 1996-12-17 Digital Equipment Corporation Order preserving data translation
US5874540A (en) * 1994-10-05 1999-02-23 Immunomedics, Inc. CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
US20030044772A1 (en) * 1997-08-04 2003-03-06 Applied Molecular Evolution [Formerly Ixsys] Methods for identifying ligand specific binding molecules
US5958442A (en) 1997-10-24 1999-09-28 Bristol-Myers Squibb Company Oncostatin M for treating inflammation
GB9806530D0 (en) * 1998-03-26 1998-05-27 Glaxo Group Ltd Inflammatory mediator
KR101077001B1 (ko) 1999-01-15 2011-10-26 제넨테크, 인크. 효과기 기능이 변화된 폴리펩티드 변이체
US20050208558A1 (en) * 1999-10-19 2005-09-22 Applera Corporation Detection kits, such as nucleic acid arrays, for detecting the expression or 10,000 or more Drosophila genes and uses thereof
US6696620B2 (en) * 2000-05-02 2004-02-24 Epicyte Pharmaceutical, Inc. Immunoglobulin binding protein arrays in eukaryotic cells
KR100857943B1 (ko) 2000-11-30 2008-09-09 메다렉스, 인코포레이티드 인간 항체의 제조를 위한 형질전환 트랜스염색체 설치류
EP1355919B1 (en) 2000-12-12 2010-11-24 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
WO2002060954A1 (en) * 2001-01-12 2002-08-08 Karolinska Innovations Ab Materials and methods for treatment of hepatitis c
US7319142B1 (en) * 2004-08-31 2008-01-15 Monsanto Technology Llc Nucleotide and amino acid sequences from Xenorhabdus and uses thereof
DK2325207T3 (en) 2004-11-12 2017-06-06 Xencor Inc Fc variants with altered binding to FcRn
US7572896B2 (en) * 2005-02-03 2009-08-11 Raven Biotechnologies, Inc. Antibodies to oncostatin M receptor
WO2006138005A2 (en) * 2005-05-10 2006-12-28 Monsanto Technology, Llc Genes and uses for plant improvement
EA035459B1 (ru) * 2005-12-29 2020-06-19 Сентокор, Инк. Антитело против il-23p19
ME00588A (en) * 2006-08-18 2011-12-20 Prlr-specific antibody and uses thereof
RS56743B1 (sr) * 2007-08-21 2018-03-30 Amgen Inc Humani c-fms antigen vezujući proteini
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
EP3825329A1 (en) * 2007-12-26 2021-05-26 Xencor, Inc. Fc variants with altered binding to fcrn
AR070315A1 (es) * 2008-02-07 2010-03-31 Merck & Co Inc Anticuerpos 1b20 antagonistas de pcsk9
US20120093833A1 (en) * 2010-10-13 2012-04-19 Juan Carlos Almagro Human Oncostatin M Antibodies and Methods of Use
PT2643352T (pt) 2010-11-23 2018-08-01 Glaxo Group Ltd Proteínas de ligação a antigénio para a oncostatina m (osm)

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070286861A1 (en) * 2004-03-30 2007-12-13 Ellis Jonathan H Immunoglobulins
US20110092372A1 (en) * 2009-10-14 2011-04-21 Juan Carlos Almagro Methods of Affinity Maturing Antibodies

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Casset et al. A peptide mimetic of an anti-CD4 monoclonal antibody by rational design. Biochemical and Biophysical Research Communications, 307:198-205, 2003. *
Colman. Effects of amino acid sequence changes on antibody-antigen interactions. Research in Immunology, 145:33-36, 1994. *
MacCallum et al. Antibody interactions: contact analysis and binding site topography. Journal of Molecular Biology, 262:732-745, 1996. *
Paul. Fv structure and diversity in three dimension. Fundamental Immunology, 3rd Edition, Raven Press, New York, Chapter 8, pages 292-295, 1993. *
Rudikoff et al. Single amino acid substitution altering antigen-binding specificity. Proceeding of the National Academy of Sciences, 79(6):1979-1983, March 1982. *
Sundberg. Structural basis of antibody-antigen interactions. Methods in Molecular Biology, 524:23-36, 2009. *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013166011A2 (en) 2012-05-02 2013-11-07 Janssen Biotech, Inc. Binding proteins having tethered light chains
US9550828B2 (en) 2013-09-05 2017-01-24 Boise State University Oncostatin M (OSM) antagonists for preventing cancer metastasis and IL-6 related disorders
US10286070B2 (en) 2013-09-05 2019-05-14 Boise State University Oncostatin M (OSM) antagonists for preventing cancer metastasis and IL-6 related disorders
US20170327573A1 (en) * 2014-09-24 2017-11-16 Universitá Degli Studi Di Padova Composition to induce bone marrow stem cell mobilization
WO2020127884A1 (fr) * 2018-12-21 2020-06-25 Universite De Poitiers PROTEINE DE LIAISON SPECIFIQUE CAPABLE DE SE LIER SPECIFIQUEMENT A L'ONCOSTATINE M HUMAINE (hOSM) ET SES UTILISATIONS
FR3090637A1 (fr) * 2018-12-21 2020-06-26 Universite De Poitiers Protéine de liaison spécifique capable de se lier spécifiquement à l’oncostatine M humaine (hOSM) et ses utilisations.
US11633457B2 (en) 2019-04-11 2023-04-25 Boise State University Pharmaceutical compositions comprising oncostatin m (OSM) antagonist derivatives and methods of use

Also Published As

Publication number Publication date
KR101920250B1 (ko) 2018-11-20
SG189322A1 (en) 2013-05-31
JP6490036B2 (ja) 2019-03-27
ZA201303429B (en) 2016-01-27
MX2013004209A (es) 2013-06-05
IL225573B (en) 2020-06-30
AR126555A2 (es) 2023-10-18
US20190092856A1 (en) 2019-03-28
JP6721655B2 (ja) 2020-07-15
TWI548749B (zh) 2016-09-11
CN103261223A (zh) 2013-08-21
EP2627356B1 (en) 2019-05-22
JO3496B1 (ar) 2020-07-05
US20160009798A1 (en) 2016-01-14
JP2019037239A (ja) 2019-03-14
IL225573A0 (en) 2013-06-27
BR112013009083A2 (pt) 2016-09-13
US10941197B2 (en) 2021-03-09
JP2013544076A (ja) 2013-12-12
EA201390493A1 (ru) 2014-02-28
EP2627356A2 (en) 2013-08-21
US20140099315A1 (en) 2014-04-10
US9587018B2 (en) 2017-03-07
US9163083B2 (en) 2015-10-20
US20170145090A1 (en) 2017-05-25
JP6216917B2 (ja) 2017-10-25
RU2013119957A (ru) 2014-11-20
KR20130099989A (ko) 2013-09-06
CA2814652C (en) 2021-05-18
US10179812B2 (en) 2019-01-15
EP2627356A4 (en) 2014-04-30
CN103261223B (zh) 2017-03-29
RU2600444C2 (ru) 2016-10-20
BR112013009083A8 (pt) 2018-01-02
TW201239094A (en) 2012-10-01
AU2011313847A1 (en) 2013-05-02
AU2011313847B2 (en) 2016-07-07
WO2012051111A2 (en) 2012-04-19
SG10201600263QA (en) 2016-02-26
CA2814652A1 (en) 2012-04-19
EA031044B1 (ru) 2018-11-30
JP2017200465A (ja) 2017-11-09
AR083418A1 (es) 2013-02-21
UA118646C2 (uk) 2019-02-25
UY33670A (es) 2012-05-31
WO2012051111A3 (en) 2012-06-21
BR112013009083B1 (pt) 2021-08-10
MX354095B (es) 2018-02-12

Similar Documents

Publication Publication Date Title
US10941197B2 (en) Method of treating osteoarthritis with human oncostatin M antibodies
US8580265B2 (en) Antibody molecules which bind IL-17A and IL-17F
US8323646B2 (en) Engineered anti-IL-13 antibodies, compositions, methods and uses
US20230287100A1 (en) Methods of Treatment Using Anti-IL-17A/F Antibodies
US20240166765A1 (en) Inflammatory disease treatment using anti-tissue factor antibodies
TW202342517A (zh) 使用抗組織因子抗體之炎性疾病治療

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE