US20130122003A1 - Methods of inhibiting tumor growth by antagonizing il-6 receptor - Google Patents

Methods of inhibiting tumor growth by antagonizing il-6 receptor Download PDF

Info

Publication number
US20130122003A1
US20130122003A1 US13/672,923 US201213672923A US2013122003A1 US 20130122003 A1 US20130122003 A1 US 20130122003A1 US 201213672923 A US201213672923 A US 201213672923A US 2013122003 A1 US2013122003 A1 US 2013122003A1
Authority
US
United States
Prior art keywords
antibody
seq
vegf
antigen
domain
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
US13/672,923
Other languages
English (en)
Inventor
Li Zhang
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.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US13/672,923 priority Critical patent/US20130122003A1/en
Publication of US20130122003A1 publication Critical patent/US20130122003A1/en
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZHANG, LI
Priority to US14/329,565 priority patent/US9409990B2/en
Priority to US15/204,639 priority patent/US20170022279A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • 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

Definitions

  • the present invention relates to compositions and methods for inhibiting or attenuating tumor growth or proliferation in a subject. More specifically, the invention relates to methods comprising administering an interleukin-6 (IL-6) antagonist to a tumor-bearing subject.
  • IL-6 interleukin-6
  • VEGF vascular endothelial growth factor
  • Interleukin-6 (“IL-6”) is a pro-inflammatory cytokine that is expressed in multiple cancer types. Clinical studies have shown that increased serum IL-6 levels are associated with worse patient outcomes. Elevated expression of IL-6 can result from the activation of oncogenic signaling pathways and/or as a consequence of chronic inflammation, which has been associated with the development of cancer. IL-6 signals through its heterodimeric receptor IL-6R/gp130 to activate the JAK/STAT and Ras signaling pathways. In particular, IL-6 strongly activates STAT3, which has been shown to promote tumor cell proliferation, invasion and survival.
  • the present invention is based, at least in part, on the surprising discovery that anti-VEGF-resistant tumors express elevated levels of IL-6 and that antagonism of IL-6 (e.g., by using an anti-IL-6R antibody), when combined with anti-VEGF therapy, is able to overcome anti-VEGF resistance in tumors and thus provides robust anti-tumor activity against tumors that heretofore were deemed unresponsive to VEGF antagonism.
  • methods and compositions are provided for inhibiting or attenuating the growth of an anti-VEGF-resistant tumor in a subject.
  • methods of cancer therapy comprising: (i) measuring the level of IL-6/STAT3 signaling in a tumor biopsy from a subject; and (ii) administering an IL-6 antagonist to the subject if the tumor biopsy exhibits increased IL-6/STAT3 signaling.
  • the methods according to this aspect of the invention also comprise administering to the subject an IL-6 antagonist and a VEGF antagonist.
  • compositions are provided which comprise at least one IL-6 antagonist and at least one VEGF antagonist.
  • methods for enhancing the anti-tumor activity of an IL-6 antagonist.
  • the methods according to this aspect of the invention comprise administering at least one additional anti-tumor agent to a tumor-bearing subject in combination with an IL-6 antagonist.
  • the additional anti-tumor agent can be, e.g.: a VEGF antagonist, an EGFR antagonist, or a combination thereof.
  • the antagonist molecules of the invention can be, e.g., antigen-specific binding proteins, including antigen-specific binding proteins that specifically bind, e.g., IL-6, IL-6R, VEGF, VEGFR1, VEGFR2, EGFR, EGFRvIII, ErbB2, ErbB3, and/or ErbB4.
  • Antigen-specific binding proteins of the present invention include antibodies and antigen-binding fragments thereof.
  • Antigen-specific binding proteins also include fusion polypeptides comprising ligand-binding portions of one or more receptor molecules.
  • the IL-6 antagonist is an antibody that specifically binds IL-6R
  • the VEGF antagonist is a VEGF-binding fusion molecule comprising VEGF binding domains of VEGFR1, VEGFR2 and a multimerizing domain (a “VEGF-Trap”)
  • the EGFR antagonist is an antibody that specifically binds EGFR (ErbB1/HER1), or an antibody that specifically binds ErbB3, or an antibody that specifically binds ErbB4.
  • VEGF-Trap VEGF-Trap
  • the EGFR antagonist is an antibody that specifically binds EGFR (ErbB1/HER1), or an antibody that specifically binds ErbB3, or an antibody that specifically binds ErbB4.
  • other antagonists can be used in the context of the present invention as described elsewhere herein.
  • FIG. 1 shows the results of Western blots on cultured A549, Calu3 and Du145 tumor cells to assess the levels of phospho-STAT3 and total STAT3 in the cells following treatment with 10 ⁇ g/ml human Fc control protein (lane 1), 10 anti-IL-6R mAb1 (lane 2), 10 ng/ml IL-6 plus 10 ⁇ g/ml hFc (lane 3), or 10 ng/ml IL-6 plus 10 ⁇ g/ml anti-IL-6R mAb1 (lane 4).
  • FIG. 2 panel A shows the results of an ELISA performed on conditioned medium collected from cultured A431—P or A431-V2 cells to measure the concentration of IL-6.
  • FIG. 2 panel B shows the results of a Western blot performed on A431-P or A431-V2 cells treated with either hFc control protein (10 ⁇ g/ml) or anti-IL-6R mAb1 (10 ⁇ g/ml), to measure the levels of phospho-STAT3 (relative to actin control).
  • the present invention provides methods for inhibiting or attenuating the growth of a tumor in a subject.
  • the invention includes administering an IL-6 antagonist to a tumor-bearing subject.
  • the IL-6 antagonist may be administered in combination with one or more additional therapeutic agents.
  • Exemplary therapeutic agents that can be administered in combination with an IL-6 antagonist, in accordance with the methods of the present invention include, e.g., antagonists of vascular endothelial growth factor (VEGF) and/or epidermal growth factor receptor (EGFR) antagonists (as defined herein). Further examples of therapeutic agents that can be administered in combination with an IL-6 antagonist in accordance with the methods of the present invention are described elsewhere herein.
  • VEGF vascular endothelial growth factor
  • EGFR epidermal growth factor receptor
  • the methods of the present invention are useful for the treatment of primary and/or metastatic tumors arising in the brain and meninges, oropharynx, lung and bronchial tree, gastrointestinal tract, male and female reproductive tract, muscle, bone, skin and appendages, connective tissue, spleen, immune system, blood forming cells and bone marrow, liver and urinary tract, and special sensory organs such as the eye.
  • Specific cancers that are treatable according to the methods of the present invention include, e.g., renal cell carcinoma, pancreatic carcinoma, breast cancer, prostate cancer, hepatocellular carcinoma, colorectal cancer, malignant mesothelioma, multiple myeloma, ovarian cancer, and melanoma.
  • an “anti-VEGF-resistant tumor,” as used herein, means a tumor that does not respond, or only partially responds, to treatment with an anti-VEGF agent such as an anti-VEGF antibody, an anti-VEGF receptor antibody, or any other VEGF-specific binding protein (including, e.g., a VEGF-trap, as defined herein).
  • an anti-VEGF agent such as an anti-VEGF antibody, an anti-VEGF receptor antibody, or any other VEGF-specific binding protein (including, e.g., a VEGF-trap, as defined herein).
  • an anti-VEGF-resistant tumor can be, e.g., a tumor that, when contacted with an amount of VEGF antagonist that is ordinarily capable of inhibiting or attenuating the growth of at least one type of tumor, continues to grow and/or proliferate in vitro or in vivo (e.g., in cell culture or when implanted into an animal).
  • An anti-VEGF-resistant tumor may be a tumor derived from tumor cells that originally responded to anti-VEGF therapy, but through selection, mutation or adaptation, have acquired resistance to one or more anti-VEGF agents.
  • the subjects that are treatable using the methods of the present invention include any subject diagnosed with cancer or identified as having a tumor.
  • the subject is a patient who has been diagnosed or identified as having a tumor that is at least partially resistant to anti-VEGF treatment.
  • Methods for diagnosing a patient as having an anti-VEGF resistant tumor will be known to persons of ordinary skill in the art and can be practiced using routine diagnostic methods.
  • the present invention also includes methods of cancer therapy comprising: (i) measuring the level of IL-6/STAT3 signaling in a subject (e.g., in a serum sample, tissue sample, tumor biopsy, etc., obtained from the subject); and (ii) administering an IL-6 antagonist to the subject if the subject (or sample/biopsy obtained therefrom) exhibits increased IL-6/STAT3 signaling.
  • the expression “increased IL 6/STAT-3 signaling” means that the amount of IL-6 and/or amount of phospho-STAT3 measured in a tumor biopsy is at least 3 ⁇ higher (e.g., 4 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ or more) than in a tumor that is sensitive (i.e., not resistant) to anti-VEGF therapy.
  • “increased IL-6/STAT-3 signaling” means that the ratio of phospho-STAT3 to an invariant control protein (e.g., the P-STAT3/actin ratio) in a sample taken from a subject is greater than about 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, or more.
  • an invariant control protein e.g., the P-STAT3/actin ratio
  • “increased IL-6/STAT-3 signaling” means that the concentration of IL-6 in a tumor sample taken from a subject is greater than about 50 pg/ml, 55 pg/ml, 60 pg/ml, 65 pg/ml, 70 pg/ml, 75 pg/ml, 80 pg/ml, 85 pg/ml, 90 pg/ml, 95 pg/ml, 100 pg/ml, 110 pg/ml, 120 pg/ml, 130 pg/ml, 140 pg/ml, 150 pg/ml, 160 pg/ml, 170 pg/ml, 180 pg/ml, 190 pg/ml, 200 pg/ml, or higher.
  • Levels of IL-6 and phospho-STAT3 can be measured, e.g., using Western blot, ELISA, or by any combination of IL-6 and
  • the present invention also includes methods for determining whether a tumor-bearing patient has an anti-VEGF-resistant tumor.
  • Methods according to this aspect of the invention comprise measuring the level of IL-6/STAT3 signaling in a sample (e.g., tumor biopsy) from a patient, wherein “increased IL-6/STAT3 signaling” in the sample (as that expression is defined herein above) identifies the subject as having an anti-VEGF-resistant tumor. Since the present inventors have demonstrated that anti-VEGF-resistant tumors are sensitive to IL-6 antagonism, the methods according to this aspect of the invention may, in certain embodiments, further comprise administering to the patient an IL-6 antagonist and/or a VEGF antagonist.
  • the present invention includes methods that comprise administering an IL-6 antagonist, a VEGF antagonist, an EGFR antagonist, and/or combinations thereof, to a subject in need thereof.
  • an “IL-6 antagonist” is any agent which binds to or interacts with IL-6 and inhibits the normal biological signaling function of IL-6 in vitro or in vivo.
  • the term “IL-6 antagonist” also includes antagonists of IL-6 receptor (“IL-6R”, i.e., “IL-6R antagonists).
  • An IL-6R antagonist may be any agent which binds to or interacts with IL-6R and inhibits the normal biological signaling function of IL-6R in vitro or in vivo.
  • a VEGF antagonist can be any agent which binds to or interacts with VEGF or a VEGF receptor (VEGFR1, also referred to as Flt1; or VEGFR2, also referred to as Flk1 or KDR).
  • VEGFR1 also referred to as Flt1
  • VEGFR2 also referred to as Flk1 or KDR
  • an EGFR antagonist can be any agent which binds to or interacts with an epidermal growth factor receptor and inhibits the normal biological signaling function of the receptor in vitro or in vivo.
  • an EGFR antagonist may be an antagonist of EGFR (also referred to as ErbB1 or HER1), an antagonist of a variant of EGFR such as, e.g., EGFRvIII, an antagonist of ErbB2 (also referred to as HER2 or Neu), an antagonist of ErbB3 (also referred to as HER3), and/or an antagonist of ErbB4 (also referred to as HER4).
  • Antagonists of IL-6, IL-6R, VEGF, VEGF receptors, and EGFRs include small molecule antagonists, as well as antigen-specific binding proteins, as described elsewhere herein.
  • the antagonists that are useful in the methods of the present invention include antigen-specific binding proteins.
  • the present invention includes methods comprising administering an antigen-specific binding protein that specifically binds interleukin-6 (IL-6) or IL-6 receptor (IL-6R) to a subject.
  • the present invention also includes methods comprising administering an antigen-specific binding protein that specifically binds vascular endothelial growth factor (VEGF), or a VEGF receptor (VEGFR), or an antigen-specific binding protein that specifically binds epidermal growth factor receptor (EGFR, EGFRvIII, ErbB2, ErbB3, and/or ErbB4) to a subject.
  • VEGF vascular endothelial growth factor
  • VEGFRvIII epidermal growth factor receptor
  • antigen-specific binding protein means a protein comprising at least one domain which specifically binds a particular antigen.
  • exemplary categories of antigen-specific binding proteins include antibodies, antigen-binding portions of antibodies, peptides that specifically interact with a particular antigen (e.g., peptibodies), receptor molecules that specifically interact with a particular antigen, and proteins comprising a ligand-binding portion of a receptor that specifically binds a particular antigen.
  • the term “specifically binds” or the like, as used herein, means that an antigen-specific binding protein, or an antigen-specific binding domain, forms a complex with a particular antigen characterized by a dissociation constant (K D ) of 500 pM or less, and/or does not bind other unrelated antigens under ordinary test conditions, “Unrelated antigens” are proteins, peptides or polypeptides that have less than 75% amino acid identity to one another. Methods for determining whether two molecules specifically bind one another are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like.
  • an antigen-specific binding protein or an antigen-specific binding domain includes molecules that bind a particular antigen (e.g., IL-6, IL-6R, VEGF, VEGFR and/or EGFR) or a portion thereof with a K D of less than about 500 pM, less than about 400 pM, less than about 300 pM, less than about 200 pM, less than about 100 pM, less than about 90 less than about 80 pM, less than about 70 pM, less than about 60 pM, less than about 50 pM, less than about 40 pM, less than about 30 pM, less than about 20 pM, less than about 10 pM, less than about 5 pM, less than about 4 pM, less than about 2 pM, less than about 1 pM, less than about 0.5 pM, less than about 0.2 pM, less than about 0.1 pM, or less than about 0.05 pM
  • a particular antigen e.g.,
  • an antigen-specific binding protein or antigen-specific binding domain “does not bind” to an unrelated antigen if the protein or binding domain, when tested for binding to the unrelated antigen at 25° C. in a surface plasmon resonance assay, exhibits a K, of greater than 1000 pM, or fails to exhibit any binding in such an assay or equivalent thereof.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcoreTM system (Biacore Life Sciences division of GE Healthcare, Piscataway, N.J.).
  • K D means the equilibrium dissociation constant of a particular protein-protein interaction (e.g., antibody-antigen interaction). Unless indicated otherwise, the K D values disclosed herein refer to K D values determined by surface plasmon resonance assay at 25° C.
  • an antigen-specific binding protein can comprise or consist of an antibody or antigen-binding fragment of an antibody that specifically binds a particular antigen (e.g., anti-IL-6 antibody, anti-IL-6R antibody, anti-VEGF antibody, anti-VEGFR antibody and/or anti-EGFR antibody, or antigen-binding fragments thereof).
  • a particular antigen e.g., anti-IL-6 antibody, anti-IL-6R antibody, anti-VEGF antibody, anti-VEGFR antibody and/or anti-EGFR antibody, or antigen-binding fragments thereof.
  • antibody includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, as well as multimers thereof (e.g., IgM).
  • Each heavy chain comprises a heavy chain variable region (abbreviated herein as HCVR or V H ) and a heavy chain constant region.
  • the heavy chain constant region comprises three domains, C H 1, C H 2 and C H 3.
  • Each light chain comprises a light chain variable region (abbreviated herein as LCVR or V L ) and a light chain constant region.
  • the light chain constant region comprises one domain (C L 1).
  • V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the FRs of the antibodies (or antigen-binding portion thereof) may be identical to the human germline sequences, or may be naturally or artificially modified.
  • An amino acid consensus sequence may be defined based on a side-by-side analysis of two or more CDRs.
  • antibody also includes antigen-binding fragments of full antibody molecules.
  • antigen-binding portion of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • Antigen-binding fragments of an antibody may be derived, e.g., from full antibody molecules using any suitable standard techniques such as proteolytic digestion or recombinant genetic engineering techniques involving the manipulation and expression of DNA encoding antibody variable and optionally constant domains.
  • DNA is known and/or is readily available from, e.g., commercial sources, DNA libraries (including, e.g., phage-antibody libraries), or can be synthesized.
  • the DNA may be sequenced and manipulated chemically or by using molecular biology techniques, for example, to arrange one or more variable and/or constant domains into a suitable configuration, or to introduce codons, create cysteine residues, modify, add or delete amino acids, etc.
  • Non-limiting examples of antigen-binding fragments include: (i) Fab fragments; (ii) F(ab′)2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments: and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide.
  • CDR complementarity determining region
  • engineered molecules such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR-grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g. monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression “antigen-binding fragment,” as used herein.
  • SMIPs small modular immunopharmaceuticals
  • an antigen-binding fragment of an antibody will typically comprise at least one variable domain.
  • the variable domain may be of any size or amino acid composition and will generally comprise at least one CDR which is adjacent to or in frame with one or more framework sequences.
  • the V H and V L domains may be situated relative to one another in any suitable arrangement.
  • the variable region may be dimeric and contain V H -V H , V H -V L or V L -V L dimers.
  • the antigen-binding fragment of an antibody may contain a monomeric V H or V L domain.
  • an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain
  • Non-limiting, exemplary configurations of variable and constant domains that may be found within an antigen-binding fragment of an antibody of the present invention include: (i) V H -C L 1; (ii) V H -C H 2; (iii) V H -C H 3; (iv) V H -C H 1-C H 2; (v) V H -C H 1-C H 2-C H 3; (vi) V H -C H 2-C H 3; (vii) V H -C L ; (viii) V L -C H 1; (ix) V L -C H 2; (x) V L -C H 3; (xi) V L -C H 1-C H 2; (xii) V L -C H 1-C H 2-C H 3; (xiii) V L -C H 2-C H 3, and (xiv) V L -
  • variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region.
  • a hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide molecule.
  • an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric V H or V L domain (e.g., by disulfide bond(s)).
  • the molecules of the present invention may comprise or consist of human antibodies and/or recombinant human antibodies, or fragments thereof.
  • human antibody includes antibodies having variable and constant regions derived from human germline immunoglobulin sequences, Human antibodies may nonetheless include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis ire vitro or by somatic mutation in viva), for example in the CDRs and in particular CDR3.
  • the term “human antibody”, as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • the molecules of the present invention may comprise or consist of recombinant human antibodies or antigen-binding fragments thereof.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further below), antibodies isolated from a recombinant, combinatorial human antibody library (described further below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor et at (1992) Nucl. Acids Res.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in viva somatic mutagenesis) and thus the amino acid sequences of the V H and V L regions of the recombinant antibodies are sequences that, while derived from and related to human germline V H and V L sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • the methods of the present invention comprises administering an anti-IL-6R antibody, or antigen-binding fragment thereof, to a subject.
  • anti-IL-6R antibody or antigen-binding fragment thereof.
  • IL-6R interleukin-6 receptor
  • the extracellular domain of human IL-6R has the amino add sequence as set forth in SEQ ID NO:1.
  • Anti-IL-6R antibodies are mentioned in, e.g., U.S. Pat. Nos. 5,795,695; 5,817,790; 6,410,691; 6,670,373; and 7,582,298.
  • anti-IL-6R antibodies mentioned and/or described in any of the foregoing publications, or antigen-binding fragments thereof, can be used in the context of the present invention.
  • a non-limiting, exemplary anti-IL-6R antibody that can be used in the context of the present invention is an anti-IL-6R antibody, or antigen-binding fragment thereof, comprising the heavy and light chain CDRs of the HCVR/LCVR amino acid pair comprising SEQ ID NOs: 2/3.
  • the anti-IL-6R antibody can be an antibody, or antigen-binding fragment thereof, comprising heavy chain CDRs (HCDR1, HCDR2 and HCDR3) having the amino acid sequences of SEQ ID NOs: 4, 5, and 6, respectively; and light chain CDRs (LCDR1, LCDR2 and LCDR3) having the amino acid sequences of SEQ ID NOs: 7, 8, and 9, respectively.
  • heavy chain CDRs HCDR1, HCDR2 and HCDR3
  • LCDR1, LCDR2 and LCDR3 having the amino acid sequences of SEQ ID NOs: 7, 8, and 9, respectively.
  • VEGF antagonist means any molecule that blocks, reduces or interferes with the normal biological activity of VEGF.
  • VEGF antagonists include molecules which interfere with the interaction between VEGF and a natural VEGF receptor, e.g., molecules which bind to VEGF or a VEGF receptor and prevent or otherwise hinder the interaction between VEGF and a VEGF receptor.
  • Specific exemplary VEGF antagonists include anti-VEGF antibodies, anti-VEGF receptor antibodies, and VEGF receptor-based chimeric molecules (also referred to herein as “VEGF-Traps”).
  • VEGF receptor-based chimeric molecules include chimeric polypeptides which comprise two or more immunoglobulin (Ig)-like domains of a VEGF receptor such as VEGFR1 (also referred to as Flt1) and/or VEGFR2 (also referred to as Flk1 or KDR), and may also contain a multimerizing domain (e.g., an Fc domain which facilitates the multimerization [e.g., dimerization] of two or more chimeric polypeptides).
  • VEGFR1 also referred to as Flt1
  • VEGFR2 also referred to as Flk1 or KDR
  • a multimerizing domain e.g., an Fc domain which facilitates the multimerization [e.g., dimerization] of two or more chimeric polypeptides.
  • An exemplary VEGF receptor-based chimeric molecule is a molecule referred to as VEGFRIR2-Fc ⁇ C1(a) which is encoded by the nucleic acid sequence of
  • VEGFR1R2-Fc ⁇ C1(a) comprises three components: (1) a VEGFR1 component comprising amino acids 27 to 129 of SEQ ID NO:11: (2) a VEGFR2 component comprising amino acids 130 to 231 of SEQ ID NO:11; and (3) a multimerization component (“Fc ⁇ C1(a)”) comprising amino acids 232 to 457 of SEQ ID NO:11 (the C-terminal amino acid of SEQ ID NO.11 [i.e., K458] may or may not be included in the VEGF antagonist used in the methods of the invention; see e.g., U.S. Pat. No. 7,396,664). Amino acids 1-26 of SEQ ID NO:11 are the signal sequence.
  • the methods of the present invention comprise administering to the subject an IL-6 antagonist in combination with one or more additional therapeutic agent(s) such as a VEGF antagonist and/or an EGFR antagonist.
  • additional therapeutic agent(s) such as a VEGF antagonist and/or an EGFR antagonist.
  • the expression “in combination with” means that the additional therapeutic agents are administered before, after, or concurrent with the IL-6 antagonist.
  • the additional therapeutic agent may be administered about 72 hours, about 60 hours, about 48 hours, about 36 hours, about 24 hours, about 12 hours, about 10 hours, about 8 hours, about 6 hours, about 4 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes or about 10 minutes prior to the administration of the IL-6 antagonist.
  • the additional therapeutic agent may be administered about 10 minutes, about 15 minutes, about 30 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours or about 72 hours after the administration of the IL-6 antagonist.
  • Administration “concurrent” with the IL-6 antagonist means that the additional therapeutic agent is administered to the subject in a separate dosage form within less than 5 minutes (before, after, or at the same time) of administration of the IL-6 antagonist, or administered to the subject as a single combined dosage formulation comprising both the additional therapeutic agent and the IL-6 antagonist (e.g., a single formulation comprising an anti-IL-6R antibody+a VEGF Trap; or a single formulation comprising an anti-IL-6R antibody+an anti-EGFR antibody; or a single formulation comprising an anti-IL-6R antibody+an anti-ErbB3 antibody; etc.).
  • a single formulation comprising an anti-IL-6R antibody+a VEGF Trap or a single formulation comprising an anti-IL-6R antibody+an anti-EGFR antibody; or a single formulation comprising an anti-IL-6R antibody+an anti-ErbB3 antibody; etc.
  • the present invention includes pharmaceutical compositions comprising an IL-6 antagonist.
  • the present invention also includes pharmaceutical compositions comprising an L-6 antagonist and a second active component such as a VEGF antagonist and/or an EGFR antagonist.
  • the present invention includes pharmaceutical compositions comprising an anti-IL-6R antibody and a VEGF-Trap molecule; the present invention also includes pharmaceutical compositions comprising an anti-IL-6R antibody and an anti-EGFR antibody.
  • Methods of treatment comprising administering such pharmaceutical compositions to a patient are also encompassed within the scope of the present invention.
  • compositions of the invention are formulated with suitable carriers, excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like.
  • suitable carriers excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like.
  • suitable carriers excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like.
  • Suitable formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. Additional suitable formulations are also described in Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311.
  • compositions of the present invention e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al., 1987, J. Biol. Chem. 262:4429-4432).
  • Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
  • infusion or bolus injection by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
  • epithelial or mucocutaneous linings e.g., oral mucosa, rectal and intestinal mucosa, etc.
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe.
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused.
  • a disposable pen delivery device there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention.
  • Examples include, but are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK), DISETRONICTM pen (Disetronic Medical Systems, Bergdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTM I, II and III (Nova Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTTM pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENTTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTM (sanofi-aventis, Frankfurt, Germany), to name only a few.
  • Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but are not limited to the SOLOSTARTM pen (sanofi-aventis), the FLEXPENTM (Novo Nordisk), and the KWIKPENTM (Eli Lilly), the SURECLICKTM Autoinjector (Amgen, Thousand Oaks, Calif.), the PENLETTM (Haselmeier, Stuttgart, Germany), the EPIPEN (Dey, L.P.), and the HUMIRATM Pen (Abbott Labs, Abbott Park Ill.), to name only a few.
  • the pharmaceutical compositions of the present invention can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:201).
  • polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), 1974, CRC Pres., Boca Raton, Fla.
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, 1984, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138). Other controlled release systems are discussed in the review by Langer, 1990, Science 249:1527-1533.
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by known methods. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of active ingredient(s) that can be administered to a subject is, generally, a therapeutically effective amount.
  • therapeutically effective amount means a dose of antigen-specific binding proteins and/or antigen-binding molecules that results in a decrease in tumor growth or weight of at least 5%, relative to a negative control, when administered to a tumor bearing animal (See, e.g., Example 1 herein).
  • a “therapeutically effective amount” of an IL-6R-specific binding protein, a VEGF-specific binding protein, and/or an EGFR-specific binding protein includes, e.g., an amount of such antigen-specific binding protein(s) that, when administered to a tumor bearing animal, causes a decrease in tumor growth or weight of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 80%, 90%, 95%, or 100%, relative to negative control-treated animals.
  • a therapeutically effective amount can be from about 0.05 mg to about 600 mg; e.g., anti-IL-6R antibodies, anti-EGFR antibodies, anti-ErbB3 antibodies, and/or VEGF-Trap molecules.
  • an anti-IL-6R antibody e.g., mAb1 discussed herein
  • the antibody may be administered to a subject at a dose of 100 mg, 150 mg, or 200 mg, (e.g., at a frequency of once a week, once every two weeks, etc.).
  • the amount of antigen-specific binding proteins of the present invention contained within the individual doses may be expressed in terms of milligrams of antibody per kilogram of patient body weight (i.e., mg/kg).
  • the anti-IL-6R antibodies, anti-EGFR antibodies, anti-ErbB3 antibodies, and/or VEGF-Trap molecules of the present invention may be administered to a patient at a dose of about 0.0001 to about 50 mg/kg of patient body weight (e.g.
  • the amount of VEGF-Trap molecule administered to the patient in a particular dose is 4 mg/kg or 6 mg/kg.
  • the active ingredients may be present in the compositions of the present invention in equal amounts, or alternatively, may be present in amounts that vary from one another by a factor of 0.5, 1, 1.5, 2, 2.5, 3, 4, 5, or more.
  • a person of ordinary skill in the art, using routine experimentation, will be able to determine the appropriate amounts of the individual components in the compositions of the present invention necessary to produce a desired therapeutic effect.
  • compositions of the present invention may be administered to a subject over a defined time course.
  • the methods according to this aspect of the invention comprise sequentially administering to a subject multiple doses of the composition(s) of the present invention.
  • sequentially administering means that each dose of the composition(s) of the present invention are administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months).
  • present invention includes methods which comprise sequentially administering to the patient an initial dose of a composition of the present invention, followed by one or more secondary doses of the composition, and optionally followed by one or more tertiary doses of the composition.
  • the terms “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration of the compositions of the present invention.
  • the “initial dose” is the dose which is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”);
  • the “secondary doses” are the doses which are administered after the initial dose;
  • the “tertiary doses” are the doses which are administered after the secondary doses.
  • the initial, secondary, and tertiary doses may all contain the same amount of active ingredient(s), but will generally differ from one another in terms of frequency of administration. In certain embodiments, however, the amount of active ingredient(s) contained in the initial, secondary and/or tertiary doses will vary from one another (e.g., adjusted up or down as appropriate) during the course of treatment.
  • each secondary and/or tertiary dose is administered 1 to 30 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more) days after the immediately preceding dose.
  • the phrase “the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose(s) of the compositions of the present invention which are administered to a subject prior to the administration of the very next dose in the sequence with no intervening doses.
  • the methods according to this aspect of the invention may comprise administering to a patient any number of secondary and/or tertiary doses of the compositions of the present invention.
  • any number of secondary and/or tertiary doses of the compositions of the present invention may comprise administering to a patient any number of secondary and/or tertiary doses of the compositions of the present invention.
  • only a single secondary dose is administered to the patient.
  • two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient.
  • only a single tertiary dose is administered to the patient.
  • two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient.
  • each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 29 days after the immediately preceding dose. Similarly, in embodiments involving multiple tertiary doses, each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 1 to 60 days after the immediately preceding dose, Alternatively, the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the treatment regimen. The frequency of administration may also be adjusted during the course of treatment by a physician depending on the needs of the individual patient following clinical examination.
  • An Anti-IL-6R Monoclonal Antibody Inhibits Tumor Xenograft Growth as a Single Agent and in Combination with Other Anti-Tumor Agents
  • anti-IL-6R antibody used in this Example, also referred to herein as “anti-IL-6R mAb1,” is an antibody comprising heavy chain CDRs (HCDR1, HCDR2 and HCDR3) having the amino acid sequences of SEQ ID NOs: 4, 5, and 6, respectively; and light chain CDRs (LCDR1, LCDR2 and LCDR3) having the amino acid sequences of SEQ ID NOs: 7, 8, and 9, respectively (i.e., the anti-IL-6R antibody designated VQ8F11-21 in U.S. Pat. No. 7,582,298).
  • HCDR1, HCDR2 and HCDR3 heavy chain CDRs having the amino acid sequences of SEQ ID NOs: 4, 5, and 6, respectively
  • light chain CDRs LCDR1, LCDR2 and LCDR3 having the amino acid sequences of SEQ ID NOs: 7, 8, and 9, respectively
  • the VEGF-Trap used in this Example is a dimer of two fusion polypeptides, each comprising: (1) a VEGFR1 component comprising amino acids 27 to 129 of SEQ ID NO:11; (2) a VEGFR2 component comprising amino acids 130 to 231 of SEQ ID NO:11; and (3) a multimerization component (“Fc ⁇ C1(a)”) comprising amino acids 232 to 457 of SEQ ID NO:11 (i.e., the VEGF-Trap designated VEGFR1R2-Fc ⁇ C1(a) in U.S. Pat. No.
  • anti-EGFR mAb2 The anti-EGFR antibody used in this Example, also referred to herein as “anti-EGFR mAb2,” is a fully human antibody generated against the extracellular domain of human EGFR/ErbB1/HER1.
  • cultured A549, Calu3 and Du145 cells were treated with: (i) 10 ⁇ g/ml human Fe control protein (“hFc”), (ii) 10 ⁇ g/ml of anti-IL-6R mAb1, (iii) 10 ng/ml IL-6 plus 10 ⁇ g/ml hFc, or (iv) 10 ng/ml of IL-6 plus 10 ⁇ g/ml anti-IL-6R mAb1.
  • Human Fc or anti-IL-6R mAb1 was added into cultured cells for 16 hours, and IL-6 was added into hFc or anti-IL-6R mAb1 pre treated cells for 30 minutes.
  • mice were administered human Fe control protein (25 mg/kg), anti-IL-6R mAb1 (25 mg/kg), VEGF-Trap (25 mg/kg) or the combination of anti-IL-6R mAb1 plus VEGF-Trap (25+25 mg/kg), All proteins were administered via subcutaneous injection twice per week. Tumor volumes were measured twice per week over the course of the experiment (on days 35, 39, 42, 46, 49, 53, 56, 60, 63 and 67 after implantation). The average tumor growth from the start of treatment was calculated for each group. The percent decrease of tumor growth was calculated from comparison to the Fc control group. The results are summarized in Table 1.
  • anti-IL-6R mAb1 an inhibitory anti-EGFR antibody
  • anti-EGFR mAb2 an inhibitory anti-EGFR antibody
  • mice were administered human Fc control protein (25 mg/kg), anti-IL-6R mAb1 (12.5 mg/kg), anti-EGFR mAb2 (12.5 mg/kg) or the combination of anti-IL-6R mAb1 plus anti-EGFR mAb2 (12.5+12.5 mg/kg). All proteins were administered via subcutaneous injection twice per week. Tumor volumes were measured twice per week over the course of the experiment (on days 34, 37, 41, 44, 48, 51, 54 and 57 after implantation) and tumor weights were determined upon excision of tumors at the conclusion of the experiment. Averages of the tumor growth from the start of treatment and the tumor weights were calculated for each group. The percent decreases of tumor growth and tumor weight were calculated from comparison to the Fc control group. The results are summarized in Tables 2 and 3.
  • anti-IL-6R mAb1 an inhibitory anti-ErbB3 antibody
  • anti-ErbB3 mAb3 an inhibitory anti-ErbB3 antibody
  • mice were administered human Fc control protein (25 mg/kg), anti-IL-6R mAb1 (12.5 mg/kg), anti-ErbB3 mAb3 (12.5 mg/kg) or the combination of anti-IL-6R mAb1 plus anti-ErbB3 mAb3 (12.5+12.5 mg/kg). All proteins were administered via subcutaneous injection twice per week. Tumor volumes were measured twice per week over the course of the experiment (on days 59, 62, 66, 69, 74, 77, 80, 83, 87 and 90 after implantation) and tumor weights were determined upon excision of tumors at the conclusion of the experiment. Averages of the tumor growth from the start of treatment and the tumor weights were calculated for each group. The percent decreases of tumor growth and tumor weight were calculated from comparison to the Fc control group. The results are summarized in Tables 4 and 5.
  • anti-IL-6R mAb1 inhibited the growth of Du145, Calu3 and A549 tumor xenografts as a single agent. Furthermore, combination treatment with anti-IL-6R mAb1 plus VEGF-Trap inhibited the growth of Du145 tumor xenografts more potently than either single agent. Also, combination treatment with anti-IL-6R mAb1 plus an inhibitory anti-EGFR antibody (anti-EGFR mAb2) inhibited the growth of Calu3 tumor xenografts more potently than either single agent.
  • anti-EGFR mAb2 an inhibitory anti-EGFR antibody
  • anti-IL-6R mAb1 preferentially inhibited the growth of tumors that exhibit autocrine IL-6/STAT3 signaling, suggesting the possibility that immunohistochemical analysis of tumor biopsies for IL-6 and/or phospho-STAT3 levels might be useful in identifying patients that are most likely to benefit from anti-IL-6R treatment.
  • anti-IL-6R mAb1 decreased phospho-STAT3 and increased cleaved caspase-3 levels in tumor xenografts, suggesting that IL-6/STAT3 signaling contributes to tumor cell survival.
  • An Anti-IL-6R Monoclonal Antibody in Combination with a VEGF Antagonist Inhibits the Growth of Anti-VEGF-Resistant Tumors
  • a variant of the A431 human epidermoid carcinoma cell line that is resistant to the effects of VEGF Trap was isolated by serial passage in the presence of VEGF-Trap in vivo.
  • the variant cell line is referred to herein as “A431-V2”, and the parental A431 cell line is referred to herein as “A431-P”.
  • the levels of phospho-STAT3 were measured in the variant and parental A431 cells. Specifically, cultured A431-P and A431-V2 cells were treated with human Fc (10 ⁇ g/ml) or anti-IL-6R mAb1 (10 ⁇ g/ml), Cell lysates were prepared and a Western blot was performed to assess the levels of phospho-STAT3. As shown in FIG. 2( b ), the basal level of phospho-STAT3 was increased in A431-V2 cells as compared to A431-P cells, and was significantly reduced by anti-IL-6R mAb1 treatment.
  • A431-V2 tumors are resistant to VEGF-Trap single agent treatment, producing only a 36% decrease in tumor growth relative to control-treated subjects. These tumors, however, were responsive to anti-IL-6R mAb1 plus VEGF-Trap combination treatment, suggesting that IL-6 contributes to the VEGF-Trap-resistant phenotype of A431-V2 tumors.
  • Preliminary data indicate that the increased IL-6 signaling in the A431-V2 tumors does not prevent the ability of VEGF-Trap to decrease tumor vascularity, suggesting that IL-6 signaling enhances the ability of tumor cells to proliferate and/or survive when the function of the tumor vasculature is impaired. This observation is consistent with the ability of anti-IL-6R mAb1 to potentiate the effect of VEGF-Trap in Du145 tumors as well (see Example 1).
  • IL-6 antagonism e.g., treatment with anti-IL-6R mAb1
  • IL-6 antagonism is a useful therapeutic strategy for treating multiple types of cancer, either as a single agent or in combination with VEGF antagonists, especially in the context of anti-VEGF-resistant tumors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US13/672,923 2011-11-10 2012-11-09 Methods of inhibiting tumor growth by antagonizing il-6 receptor Abandoned US20130122003A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/672,923 US20130122003A1 (en) 2011-11-10 2012-11-09 Methods of inhibiting tumor growth by antagonizing il-6 receptor
US14/329,565 US9409990B2 (en) 2011-11-10 2014-07-11 Methods of inhibiting tumor growth by antagonizing IL-6 receptor
US15/204,639 US20170022279A1 (en) 2011-11-10 2016-07-07 Methods of inhibiting tumor growth by antagonizing il-6 receptor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161557939P 2011-11-10 2011-11-10
US201261609968P 2012-03-13 2012-03-13
US201261613538P 2012-03-21 2012-03-21
US13/672,923 US20130122003A1 (en) 2011-11-10 2012-11-09 Methods of inhibiting tumor growth by antagonizing il-6 receptor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/329,565 Continuation US9409990B2 (en) 2011-11-10 2014-07-11 Methods of inhibiting tumor growth by antagonizing IL-6 receptor

Publications (1)

Publication Number Publication Date
US20130122003A1 true US20130122003A1 (en) 2013-05-16

Family

ID=47179020

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/672,923 Abandoned US20130122003A1 (en) 2011-11-10 2012-11-09 Methods of inhibiting tumor growth by antagonizing il-6 receptor
US14/329,565 Active US9409990B2 (en) 2011-11-10 2014-07-11 Methods of inhibiting tumor growth by antagonizing IL-6 receptor
US15/204,639 Abandoned US20170022279A1 (en) 2011-11-10 2016-07-07 Methods of inhibiting tumor growth by antagonizing il-6 receptor

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/329,565 Active US9409990B2 (en) 2011-11-10 2014-07-11 Methods of inhibiting tumor growth by antagonizing IL-6 receptor
US15/204,639 Abandoned US20170022279A1 (en) 2011-11-10 2016-07-07 Methods of inhibiting tumor growth by antagonizing il-6 receptor

Country Status (20)

Country Link
US (3) US20130122003A1 (ru)
EP (1) EP2776468B1 (ru)
JP (3) JP6144689B2 (ru)
KR (1) KR101718748B1 (ru)
CN (1) CN103930443B (ru)
AR (1) AR088671A1 (ru)
AU (2) AU2012335596B2 (ru)
BR (1) BR112014011211A2 (ru)
CA (1) CA2853836C (ru)
IL (2) IL231854A (ru)
IN (1) IN2014CN02923A (ru)
JO (1) JO3370B1 (ru)
MX (1) MX357837B (ru)
MY (1) MY163953A (ru)
RU (1) RU2648150C2 (ru)
SG (1) SG11201401286QA (ru)
TW (3) TWI604852B (ru)
UY (1) UY34444A (ru)
WO (1) WO2013071016A2 (ru)
ZA (1) ZA201402414B (ru)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
DE202014010421U1 (de) 2013-12-17 2015-11-12 Kymab Limited Menschliche Ziele
WO2020146239A1 (en) * 2019-01-07 2020-07-16 Celledit Llc Modified immune cells co-expressing chimeric antigen receptor and il-6 antagonist for reducing toxicity and uses thereof in adoptive cell therapy
EP3758737A4 (en) * 2018-03-02 2022-10-12 Kodiak Sciences Inc. IL-6 ANTIBODIES AND FUSION CONSTRUCTS AND CONJUGATES THEREOF
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder
US11965176B2 (en) 2018-03-14 2024-04-23 Hunan Siweikang Therapeutics Co. Ltd Modification of immune cells for reducing toxicity and uses thereof in adoptive cell therapy

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JO3370B1 (ar) * 2011-11-10 2019-03-13 Regeneron Pharma طريقة لتثبيط نمو الورم عن طريق تثبيط مستقبل انترلوكين 6
WO2015000181A1 (zh) * 2013-07-05 2015-01-08 华博生物医药技术(上海)有限公司 新型重组融合蛋白及其制法和用途
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
CA2953698A1 (en) * 2014-06-28 2015-12-30 Kodiak Sciences Inc. Dual pdgf/vegf antagonists
WO2016196377A1 (en) * 2015-05-29 2016-12-08 Merrimack Pharmaceuticals, Inc. Combination cancer therapies
AU2016381964B2 (en) 2015-12-30 2024-02-15 Kodiak Sciences Inc. Antibodies and conjugates thereof
US20180206726A1 (en) 2016-12-07 2018-07-26 Progenity Inc. Gastrointestinal tract detection methods, devices and systems
WO2018112237A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with an il-6r inhibitor
CN108567981A (zh) * 2018-07-17 2018-09-25 漯河医学高等专科学校 白细胞介素-6的拮抗剂在制备抑制Notch-1蛋白表达、治疗胰腺癌的药物中的应用
EP3883635A1 (en) 2018-11-19 2021-09-29 Progenity, Inc. Methods and devices for treating a disease with biotherapeutics
CN115666704A (zh) 2019-12-13 2023-01-31 比奥拉治疗股份有限公司 用于将治疗剂递送至胃肠道的可摄取装置

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7396664B2 (en) * 1999-06-08 2008-07-08 Regeneron Pharmaceuticals, Inc. VEGF-binding fusion proteins and nucleic acids encoding the same

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5171840A (en) 1988-01-22 1992-12-15 Tadamitsu Kishimoto Receptor protein for human B cell stimulatory factor-2
US5670373A (en) 1988-01-22 1997-09-23 Kishimoto; Tadamitsu Antibody to human interleukin-6 receptor
DE69229482T2 (de) 1991-04-25 1999-11-18 Chugai Seiyaku K.K., Tokio/Tokyo Rekombinierte humane antikörper gegen den humanen interleukin 6-rezeptor
US5888510A (en) 1993-07-21 1999-03-30 Chugai Seiyaku Kabushiki Kaisha Chronic rheumatoid arthritis therapy containing IL-6 antagonist as effective component
EP0800829B2 (en) 1994-12-29 2012-07-25 Chugai Seiyaku Kabushiki Kaisha Use of a pm-1 antibody or of a mh 166 antibody for enhancing the anti-tumor effect of cisplatin or carboplatin
US5795695A (en) 1996-09-30 1998-08-18 Xerox Corporation Recording and backing sheets containing linear and cross-linked polyester resins
BRPI0011407B8 (pt) * 1999-06-08 2021-05-25 Regeneron Pharma molécula de ácido nucléico quimérica, polipeptídeo de fusão, vetor de expressão, sistema de vetor hospedeiro, e, métodos de produzir um polipeptídeo de fusão
US7087411B2 (en) * 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
ATE316531T1 (de) 1999-10-07 2006-02-15 Lilly Co Eli Kondensierte dihydrochinolinon-derivate zur hemmung von mrp1
WO2002036164A1 (fr) 2000-10-27 2002-05-10 Chugai Seiyaku Kabushiki Kaisha Agent abaissant le taux sanguin de vegf contenant un antagoniste de il-6 en tant que principe actif
US7595378B2 (en) * 2001-06-13 2009-09-29 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
PT1639013E (pt) * 2003-07-02 2012-12-03 Innate Pharma Anticorpos contra receptor nk pan-kir2dl e a sua utilização em diagnóstico e terapêutica
TWI350175B (en) 2003-10-17 2011-10-11 Chugai Pharmaceutical Co Ltd Mesothelioma therapeutic agent
EP1967209B1 (en) 2005-11-25 2012-06-06 Keio University Therapeutic agent for prostate cancer
US8080248B2 (en) 2006-06-02 2011-12-20 Regeneron Pharmaceuticals, Inc. Method of treating rheumatoid arthritis with an IL-6R antibody
MY159787A (en) * 2006-06-02 2017-01-31 Regeneron Pharma High affinity antibodies to human il-6 receptor
WO2008154927A1 (en) * 2007-06-21 2008-12-24 Genmab A/S Novel methods for treating egfr-associated tumors
CN102438653B (zh) * 2008-11-25 2019-04-23 奥尔德生物制药公司 预防或治疗恶病质、虚弱、疲劳和/或发烧il-6拮抗剂
US8323649B2 (en) * 2008-11-25 2012-12-04 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
JO3417B1 (ar) * 2010-01-08 2019-10-20 Regeneron Pharma الصيغ المستقرة التي تحتوي على الأجسام المضادة لمضاد مستقبل( interleukin-6 (il-6r
JO3370B1 (ar) * 2011-11-10 2019-03-13 Regeneron Pharma طريقة لتثبيط نمو الورم عن طريق تثبيط مستقبل انترلوكين 6

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7396664B2 (en) * 1999-06-08 2008-07-08 Regeneron Pharmaceuticals, Inc. VEGF-binding fusion proteins and nucleic acids encoding the same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Dalwadi et al, Clinical Cancer Research, 2005, Vol. 11, pages 7674-7682. *
Tabernero, Molecular Cancer Research, 2007, Vol. 5, No. 3, pages 203-220. *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE202014010421U1 (de) 2013-12-17 2015-11-12 Kymab Limited Menschliche Ziele
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
EP3758737A4 (en) * 2018-03-02 2022-10-12 Kodiak Sciences Inc. IL-6 ANTIBODIES AND FUSION CONSTRUCTS AND CONJUGATES THEREOF
US12071476B2 (en) 2018-03-02 2024-08-27 Kodiak Sciences Inc. IL-6 antibodies and fusion constructs and conjugates thereof
US11965176B2 (en) 2018-03-14 2024-04-23 Hunan Siweikang Therapeutics Co. Ltd Modification of immune cells for reducing toxicity and uses thereof in adoptive cell therapy
WO2020146239A1 (en) * 2019-01-07 2020-07-16 Celledit Llc Modified immune cells co-expressing chimeric antigen receptor and il-6 antagonist for reducing toxicity and uses thereof in adoptive cell therapy
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Also Published As

Publication number Publication date
RU2648150C2 (ru) 2018-03-22
MX2014004886A (es) 2014-07-09
BR112014011211A2 (pt) 2017-04-25
JO3370B1 (ar) 2019-03-13
US20140322215A1 (en) 2014-10-30
CN103930443B (zh) 2017-08-04
MY163953A (en) 2017-11-15
WO2013071016A2 (en) 2013-05-16
MX357837B (es) 2018-07-26
WO2013071016A3 (en) 2013-07-25
KR101718748B1 (ko) 2017-03-22
IL248620A0 (en) 2016-12-29
US9409990B2 (en) 2016-08-09
TW201332569A (zh) 2013-08-16
CA2853836C (en) 2019-07-23
TWI604852B (zh) 2017-11-11
AU2018203666A1 (en) 2018-06-14
AU2012335596A1 (en) 2014-05-22
JP2014534237A (ja) 2014-12-18
CA2853836A1 (en) 2013-05-16
US20170022279A1 (en) 2017-01-26
JP2018083841A (ja) 2018-05-31
TW201716776A (zh) 2017-05-16
KR20140095481A (ko) 2014-08-01
RU2014118739A (ru) 2015-12-20
UY34444A (es) 2013-05-31
IL231854A (en) 2016-11-30
ZA201402414B (en) 2015-03-25
CN103930443A (zh) 2014-07-16
IL231854A0 (en) 2014-05-28
SG11201401286QA (en) 2014-05-29
TW201806620A (zh) 2018-03-01
AU2012335596B2 (en) 2018-05-17
JP2017095489A (ja) 2017-06-01
JP6144689B2 (ja) 2017-06-07
EP2776468B1 (en) 2018-01-24
EP2776468A2 (en) 2014-09-17
JP6283754B2 (ja) 2018-02-21
IN2014CN02923A (ru) 2015-07-03
AR088671A1 (es) 2014-06-25

Similar Documents

Publication Publication Date Title
US9409990B2 (en) Methods of inhibiting tumor growth by antagonizing IL-6 receptor
US11655291B2 (en) Compositions and methods for increasing muscle mass and muscle strength by specifically antagonizing GDF8 and or activin A
JP6486268B2 (ja) 抗egfr抗体及びその使用
EP2459592B1 (en) High affinity human antibodies to human angiopoietin-2
US11248044B2 (en) Methods for altering body composition by administering a GDF8 inhibitor and an Activin A inhibitor

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ZHANG, LI;REEL/FRAME:030734/0326

Effective date: 20130703

STCB Information on status: application discontinuation

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