US20140065142A1 - Multivalent and Monovalent Multispecific Complexes and Their Uses - Google Patents

Multivalent and Monovalent Multispecific Complexes and Their Uses Download PDF

Info

Publication number
US20140065142A1
US20140065142A1 US13/838,438 US201313838438A US2014065142A1 US 20140065142 A1 US20140065142 A1 US 20140065142A1 US 201313838438 A US201313838438 A US 201313838438A US 2014065142 A1 US2014065142 A1 US 2014065142A1
Authority
US
United States
Prior art keywords
antibody
mrd
target
multivalent
binding
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
US13/838,438
Other languages
English (en)
Inventor
Viktor Roschke
David LaFleur
David M. Hilbert
Peter Kiener
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.)
Zyngenia Inc
Original Assignee
Zyngenia 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 Zyngenia Inc filed Critical Zyngenia Inc
Priority to US13/838,438 priority Critical patent/US20140065142A1/en
Publication of US20140065142A1 publication Critical patent/US20140065142A1/en
Priority to US14/481,181 priority patent/US20150210765A1/en
Priority to US15/458,079 priority patent/US10526381B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • 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
    • C07K16/241Tumor Necrosis Factors
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • C07K16/2848Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily against integrin beta3-subunit-containing molecules, e.g. CD41, CD51, CD61
    • 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
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/35Fusion polypeptide containing a fusion for enhanced stability/folding during expression, e.g. fusions with chaperones or thioredoxin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids

Definitions

  • This invention relates generally to compositions containing multivalent multispecific complexes and to compositions containing multivalent and monovalent multispecific complexes having scaffolds, such as antibodies, that support such binding functionalities.
  • the invention also generally relates to methods of making these multispecific compositions and the diagnostic and therapeutic uses of these compositions.
  • bispecific or multi-specific molecules that target two or more targets simultaneously offers a novel and promising solution for discovering new systems-oriented multitargeted agents demonstrating improved efficacy and pharmacological properties over conventional monotherapies.
  • Numerous attempts to develop multispecific molecules have been based on immunoglobulin-like domains or subdomains.
  • bispecific antibodies have been prepared by chemically linking two different monoclonal antibodies or by fusing two hybridoma cell lines to produce a hybrid-hybridoma.
  • immunoglobulin-like domain-based technologies that have created multispecific, and/or multivalent molecules include dAbs, diabodies, TandAbs, nanobodies, BiTEs, SMIPs, DNLs, Affibodies, Fynorners, Kunitz Domains, Albu-dabs, DARTs, DVD-IG, Covx-bodies, peptibodies, scFv-Igs, SVD-Igs, dAb-Igs, Knobs-in-Holes, DuoBodiesTM and triomAbs.
  • each of these molecules may bind one or more targets, they each present challenges with respect to retention of typical Ig function (e.g., half-life, effector function), production (e.g., yield, purity), valency, simultaneous target recognition, and bioavailability.
  • typical Ig function e.g., half-life, effector function
  • production e.g., yield, purity
  • valency e.g., simultaneous target recognition, and bioavailability.
  • VASP polypeptides based VASP polypeptides, Avian pancreatic polypeptide (aPP), Tetranectin (based on CTLD3), Affilin (based on ⁇ B-crystallin/ubiquitin) knottins, SH3 domains, PDZ domains, Tendamistat, Transferrin, an ankyrin consensus repeat domain (e.g., DARPins), lipocalin protein folds (e.g., Duocalins), fibronectin (see for example, US Application Publ. Nos.
  • the invention relates to compositions containing multivalent as well as multivalent and monovalent, multispecific complexes having scaffolds, such as, antibodies, that support such binding functionalities.
  • the invention is based in part on the surprising discovery that multispecific and multivalent binding compositions, such as those generated using the ZYBODYTM platform (Zyngenia, Inc.; see, e.g., Intl. Pub. No. WO 2009/088805 which is herein incorporated by reference) demonstrate dramatic synergistic biological activity compared to conventional monotherapy combinations.
  • This synergistic activity is expected to extend to novel therapies, for treating or preventing cancer, diseases or disorders of the immune system (e.g., autoimmune diseases such as, rheumatoid arthritis, and IBD), skeletal system (e.g., osteoporosis), cardiovascular system (e.g., stroke, heart disease), nervous system (e.g., Alzheimer's), infectious disease (e.g., HIV), and other diseases or disorders described herein or otherwise known in the art.
  • diseases or disorders of the immune system e.g., autoimmune diseases such as, rheumatoid arthritis, and IBD
  • skeletal system e.g., osteoporosis
  • cardiovascular system e.g., stroke, heart disease
  • nervous system e.g., Alzheimer's
  • infectious disease e.g., HIV
  • the invention is directed to treating a disease or disorder by administering a therapeutically effective amount of a multivalent and monovalent multispecific composition to a patient in need thereof. In a further embodiment, the invention is directed to treating a disease or disorder by administering a therapeutically effective amount of a multivalent and multispecific MRD-containing antibody to a patient in need thereof.
  • the multivalent and monovalent multispecific composition contains 2 binding sites for three or more targets. In an additional embodiment, the multivalent and monovalent multispecific composition contains 2 binding sites for four or more targets. In another embodiment, the multivalent and monovalent multispecific composition contains 2 binding sites for five or more targets. According to some embodiments, at least 1, 2, 3, 4 or more of the targets are located on a cell surface. According to some embodiments, at least 1, 2, 3, 4 or more of the targets are soluble targets (e.g., chemokines, cytokines, and growth factors). In additional embodiments, the multivalent and monovalent multispecific composition binds 1, 2, 3, 4 or more of the targets described herein.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with cancer. In a further embodiment the targets bound by the multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4 or more different signaling pathways or modes of action associated with cancer.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system. In a further embodiment the targets bound by the multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4 or more different signaling, pathways or modes of action associated with a disease or disorder of the immune system.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4, 5 or more different signaling pathways or modes of action associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • the multivalent and monovalent multispecific composition binds at least 1, 2, 3, 4, 5 or more of the targets described herein.
  • the multivalent and monovalent multispecific composition contains 2 binding sites for three or more targets. In an additional embodiment, the multivalent and monovalent multispecific composition contains 2 binding sites for four or more targets. In an additional embodiment, the multivalent and monovalent multispecific composition contains 2 binding sites for five or more targets.
  • the multivalent and monovalent multispecific composition contains 2 binding sites for three or more targets. In an additional embodiment, the multivalent and monovalent multispecific composition contains 2 binding sites for four or more targets. In another embodiment, the multivalent and monovalent multispecific composition contains 2 binding sites for five or more targets. According to some embodiments, at least 1, 2, 3, 4, or more of the targets are associated with the cell membrane. According to some embodiments, at least 1, 2, 3, 4, or more of the targets are soluble targets (e.g., chemokines, cytokines, and growth factors). In additional embodiments, the multivalent and monovalent multispecific composition binds 1, 2, 3, 4, or more of the targets described herein.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with cancer. In a further embodiment the targets bound by the multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4, or more different signaling pathways or modes of action associated with cancer.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system.
  • the targets bound by the-multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4, or more different signaling pathways or modes of action associated with a disease or disorder of the immune system.
  • the multivalent and monovalent multispecific composition binds (1) a target on a cell or tissue of interest (e.g., a tumor associated antigen on a tumor cell, an immune cell, a diseased cell or an infectious agent) and (2) a target on an effector cell.
  • a target on a cell or tissue of interest e.g., a tumor associated antigen on a tumor cell, an immune cell, a diseased cell or an infectious agent
  • the binding of one or more targets by the multivalent and monovalent multispecific composition directs an immune response to a cell, tissue, infectious agent, or other location of interest in a patient.
  • the effector cell is a leukocyte, such as a T cell or natural killer cell.
  • the effector cell is an accessory cell, such as a myeloid cell or a dendritic cell.
  • the multivalent and monovalent multispecific composition binds (1) a target on a cell or tissue of interest (e.g., a tumor associated antigen on a tumor cell, an immune cell, a diseased cell or an infectious agent) and (2) a target on a leukocyte, such as a T-cell receptor molecule.
  • a target on a cell or tissue of interest e.g., a tumor associated antigen on a tumor cell, an immune cell, a diseased cell or an infectious agent
  • a target on a leukocyte such as a T-cell receptor molecule.
  • the binding of one or more targets by the multivalent and monovalent multispecific composition directs an immune response to an infectious agent, cell, tissue, or other location of interest in a patient.
  • the multivalent and monovalent multispecific composition binds a target on the surface of a T cell.
  • the composition binds a CD3 target selected from CD3 delta, CD3 epsilon, CD3 gamma, CD3 zeta, TCR alpha, TCR beta, and multimers of proteins in the CD3 (TCR) complex.
  • the multivalent and monovalent multispecific composition binds CD3.
  • the multivalent and monovalent multispecific composition binds CD2.
  • the multivalent and monovalent multispecific composition binds a target expressed on a natural killer cell.
  • the multivalent and monovalent multispecific composition binds a target selected from: CD2, CD56, and CD161.
  • the multivalent and monovalent multispecific composition hinds a target expressed on an accessory (e.g., myeloid) cell.
  • the multivalent and monovalent multispecific composition binds a target selected from: CD64 (i.e., Fc gamma RI), an MHC class 2 and its invariant chain, TLR1, TLR2, TLR4, TLR5, and TLR6.
  • the multivalent and monovalent multispecific composition (e.g., an MRD containing antibody) has a single binding site (i.e., is monovalent) for a target.
  • the multivalent and monovalent multispecific composition has a single binding site for a target on a leukocyte, such as a T-cell (e.g., CD3), and multiple binding sites (i.e., is multivalent) for a target on, a cell or tissue of interest (e.g., a tumor associated antigen on a tumor cell, such as a target disclosed herein).
  • the multispecific composition contains single binding sites for 2 different targets (i.e., monovalently binds more than one different target).
  • the cell or tissue of interested is a cancer cell, immune cell, diseased cell, or an infectious agent.
  • a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) has a single binding site for CD3.
  • the multivalent and monovalent multispecific composition has a single binding site for CD3 and multiple binding sites for 1, 2, 3, 4, 5 or more different targets (e.g., a tumor antigen or other target disclosed herein).
  • the multispecific composition has a single binding site for CD3 and a single binding site for a different target (i.e., monovalently binds CD3 and a different target).
  • a multivalent and monovalent multispecific composition has a single binding site for CD3 epsilon.
  • the multivalent and monovalent multispecific composition has a single binding site for CD3 epsilon and multiple binding sites for 1, 2, 3, 4, 5 or more different targets (e.g., a tumor antigen or other target disclosed herein).
  • the multispecific composition has a single binding site for CD3 epsilon and a single binding site for a different target (i.e., monovalently binds CD3 epsilon and a different target).
  • the multivalent and monovalent multispecific composition has multiple binding sites for a target on a cancer cell selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • a cancer cell selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • the invention is directed to treating a disease or disorder by administering a therapeutically effective amount of a multivalent and monovalent multispecific composition that has a single binding site for a target (i.e., that monovalently binds a target) to a patient in need thereof.
  • a target i.e., that monovalently binds a target
  • the administered multivalent and monovalent multispecific composition has a single binding site for a target on a leukocyte such as a T-cell (e.g., CD3).
  • the administered multivalent and monovalent multispecific composition has a single binding site for a target on a leukocyte such as a T-cell (e.g., CD3) and multiple binding sites for (i.e., is capable of multivalently binding) a target located on a cell or tissue of interest (e.g., a tumor antigen on a tumor cell).
  • a leukocyte such as a T-cell (e.g., CD3)
  • multiple binding sites for i.e., is capable of multivalently binding
  • a target located on a cell or tissue of interest e.g., a tumor antigen on a tumor cell.
  • the multispecific composition has a single binding site for a target on a leukocyte (e.g., CD3) and a single binding site for a different target.
  • the cell of interest is a tumor cell from a cancer selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • the multivalent and monovalent multispecific composition has multiple binding sites for a target on a neurological tumor.
  • the neurological tumor is a glioma (e.g., a glioblastoma, glioblastoma multiforme (GBM), and astrocytoma), ependymoma, oligodendroglioma, neurofibroma, sarcoma, medulloblastoma, primitive neuroectodermal tumor, pituitary adenoma, neuroblastoma or cancer of the meninges (e.g., meningioma, meningiosarcoma and gliomatosis).
  • a glioma e.g., a glioblastoma, glioblastoma multiforme (GBM), and astrocytoma
  • ependymoma e.g., a glioblastoma, glioblastoma multiforme (GBM), and astrocytoma
  • ependymoma
  • the invention is directed to treating a disease or disorder by administering to a patient in need thereof, a therapeutically effective amount of a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that has a single binding site for a target (i.e., that monovalently binds a target) and multiple binding sites for 1, 2, 3, 4, 5 or more different targets.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • the multivalent and monovalent multispecific composition has single binding sites for 2 different targets.
  • the multivalent and monovalent multispecific composition has multiple binding sites for a target on a cancer cell selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • a cancer cell selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • the invention is directed to treating a disease or disorder by administering to a patient in need thereof, a therapeutically effective amount of a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that has a single binding site for CD3 (e.g., CD3 epsilon) that monovalently binds CD3 and multiple binding sites for 1, 2, 3, 4, 5 or more different targets located on a cell or tissue of interest (e.g., a tumor antigen on a tumor cell).
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • CD3 e.g., CD3 epsilon
  • targets located on a cell or tissue of interest e.g., a tumor antigen on a tumor cell.
  • the administered multivalent and monovalent multispecific composition has a single binding site for CD3 (e.g., CD3 epsilon) and a single binding site for a different target and also has multiple binding sites for a target located on a cell or tissue of interest (e.g., a tumor antigen on a tumor cell).
  • the multivalent and monovalent multispecific composition has multiple binding sites for a target on a cancer cell selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) has a single binding site for (i.e., monovalently binds) a cell surface target that requires multimerization for signaling.
  • the multivalent and monovalent multispecific composition has a single binding site for a growth factor receptor.
  • the multivalent and monovalent multispecific composition has a single binding sire for a TNF receptor superfamily member.
  • the multispecific composition additionally has a single binding site for a different target i.e., monovalently binds more than one different target).
  • the multivalent and monovalent multispecific composition binds a target associated with an endogenous blood brain barrier (BBB) receptor mediated transport system and is capable of crossing to the brain (cerebrospinal fluid) side of the BBB.
  • BBB blood brain barrier
  • the multivalent and monovalent multispecific composition has two or more binding sites for a target antigen associated with an endogenous BBB receptor mediated transport system.
  • the multivalent and monovalent multispecific composition has a single binding site for a tai get associated with an endogenous BBB receptor mediated transport system (e.g., the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and the IGF receptor mediated transport systems).
  • the multivalent and monovalent multispecific composition additionally binds 1, 2, 3, 4, 5, or more targets located on, the brain side of the BBB.
  • the MRD-containing antibody binds 1, 2, 3, 4, 5, or more targets associated with a neurological disease or disorder.
  • the multivalent and monovalent multispecific composition is administered to a patient to treat a brain cancer, metastatic cancer of the brain, or primary cancer of the brain.
  • the multivalent and monovalent multispecific composition is administered to a patient to treat brain injury, stroke, spinal cord injury, or to manage pain.
  • targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • a targets bound by the multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4, 5 or more different signaling pathways or modes of action associated with one or more of the above diseases or disorders.
  • the multivalent and monovalent multispecific composition binds 1, 2, 3, 4, 5 or more of the targets described herein.
  • the multivalent and monovalent multispecific composition is a ZYBODYTM (referred to herein as an “MRD-containing antibody,” or the like).
  • MRD-containing antibody contains binding sites for three or more targets.
  • the MRD-containing antibody contains 2 binding sites for four or more targets.
  • the MRD-containing antibody contains 2 binding sites for five or more targets.
  • the multivalent and monovalent multispecific composition contains 2 binding sites for three or more targets.
  • the multispecific composition e.g., MRD-containing antibody
  • the multispecific composition e.g., MRD-containing antibody
  • at least 1, 2, 3, 4 or more of the targets are located on a cell surface.
  • at least 1, 2, 3, 4 or more of the targets are soluble targets (e.g., chemokines, cytokines, and growth factors).
  • the MRD-containing antibody binds at least 1, 2, 3, 4, 5 or more of the targets described herein.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with cancer.
  • the targets bound by MRD-containing antibody are associated with 1, 2, 3, 4 or more different signaling pathways or modes of action associated with cancer.
  • a target bound by the multivalent and monovalent multispecific composition is associated with a disease or disorder of the immune system.
  • the targets bound by the MRD-containing antibody are associated with 1, 2, 3, 4, 5 or more different signaling pathways or modes of action associated with a disease or disorder of the immune system.
  • a target bound by the multivalent and monovalent multispecific composition is associated with a disease or disorder of the skeletal system, cardiovascular system, nervous system, or an infectious disease.
  • a target bound by the MRD-containing antibody is associated with 1, 2, 3, 4 or more different signaling pathways or modes of action associated with one or more of the above diseases or disorders.
  • the MRD-containing antibody binds 1, 2, 3, 4 or more of the targets described herein.
  • the multivalent and multispecific compositions of the invention provide the ability to selectively target multiple targets (e.g., receptors and microenvironment associated targets) having for example, different, overlapping, or redundant mechanisms of action associated with the etiology or pathophysiology of a disease or disorder.
  • targets e.g., receptors and microenvironment associated targets
  • the invention encompasses a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that is covalently or otherwise associated with a cytotoxic agent.
  • the cytoxic agent is covalently attached to an MRD-containing antibody by a linker.
  • the cytotoxic agent is a chemotherapeutic agent, growth inhibitory agent, toxin (e.g., an enzymatically active toxin of bacterial, fangal, plant, or animal origin, or fragments thereof), radioactive isotope (i.e., a radioconjugate), or prodrug.
  • the compositions of the invention are optionally linked to the cytotoxic agent by a linker.
  • a linker attaching the multivalent and monovalent multispecific composition and the cytotoxic agent is cleavable by a protease.
  • a linker attaching the multivalent and monovalent multispecific composition and the cytotoxic agent is cleavable under low pH or reducing conditions.
  • the multivalent and multispecific compositions is covalently or otherwise associated with a cytotoxic agent selected from, for example, a toxin, a chemotherapeutic agent, a drug moiety (e.g., a chemotherapeutic agent or prodrug), an antibiotic, a radioactive isotope, a chelating ligand DOTA, DOTP, DOTMA, DTPA and TETA), and a nucleolytic enzyme.
  • a cytotoxic agent selected from, for example, a toxin, a chemotherapeutic agent, a drug moiety (e.g., a chemotherapeutic agent or prodrug), an antibiotic, a radioactive isotope, a chelating ligand DOTA, DOTP, DOTMA, DTPA and TETA), and a nucleolytic enzyme.
  • the cytotoxic agent is selected from auristatin and dolostantin, MMAE, MMAF, and a maytansinoid derivative (e.g., the DM1 (N(2′)-deacetyl-N (2′)-(3-mercapto-1-oxopropyl)-maytansine), DM3 (N(2′)-deacetyl-N2-(4-mercapto-1-oxopentyl)-maytansine), and DM4 (N(2′)-deacetyl-N2-(4-mercapto-4-methyl-1-oxopentyl)-maytansine).
  • DM1 N(2′)-deacetyl-N (2′)-(3-mercapto-1-oxopropyl)-maytansine
  • DM3 N(2′)-deacetyl-N2-(4-mercapto-1-oxopentyl)-maytansine
  • DM4
  • a multivalent and monovalent multispecific composition of the invention is administered in combination with a multitargeting therapeutic.
  • a multivalent and monovalent muitispecific composition is administered in combination with a multitargeting protein kinase inhibitor.
  • a multivalent and monovalent multispecific composition is administered in combination with an NFKB inhibitor.
  • a multivalent and monovalent multispecific composition is administered in combination with an HDAC inhibitor.
  • a multivalent and monovalent multispecific composition is administered in combination with an HSP70 or HSP90 inhibitor.
  • a multivalent and monovalent multispecific composition is administered in combination with chemotherapy.
  • a multivalent and monovalent multispecific composition of the invention e.g., an MRD-containing antibody
  • a monospecific therapeutic e.g., a monoclonal antibody
  • a multivalent and monovalent multispecific composition of the invention is a full-length antibody comprising at least one modular recognition domain (MRD).
  • the full-length antibody comprises multiple MRDs.
  • the full-length antibody comprises more than one type of MRD (i.e., multiple MRDs having the same or different specificities). Also embodied in the present invention are variants and derivatives of such antibody complexes.
  • the MRDs of the MRD containing antibodies can be operably attached to the antibodies at any location on the antibody (e.g., the amino terminus of the heavy chain or light chain or the carboxyl terminus of the heavy chain or light chain), can be linked at the same or different termini, and are optionally operably linked to one another or to the antibody by a linker.
  • the antibodies of the MRD containing antibodies can be any immunoglobulin molecule that binds to an antigen and can be of any type, class, or subclass. In some embodiments, the antibody is humanized or human. In other embodiments, the antibodies also include modifications that do not interfere with their ability to bind antigen.
  • the multivalent and multispecific compositions include modifications that increase ADCC, decrease ADCC, increase CDC, or decrease CDC, that increase antibody half-life, or decrease antibody half-life compared to the antibody without the modification.
  • the antibodies of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) of the invention can be any antibody that binds to a target of therapeutic or diagnostic value.
  • the antibody of the MRD-containing antibody binds to a validated target.
  • the antibodies corresponding to the MRD containing antibodies are in clinical trials for regulatory approval.
  • the antibodies corresponding to the MRD containing antibodies are marketed.
  • the antibody binds to a cell surface antigen. In another embodiment, the antibody binds to an angiogenic factor. In a further embodiment, the antibody binds to an angiogenic receptor.
  • the antibody of the MRD-containing antibody binds to a target selected from: EGFR, ErbB2, ErbB3, ErbB4, CD20, insulin-like growth factor-I receptor, VEGF, VEGF-R and prostate specific membrane antigen.
  • the antibody of the MRD-containing antibody binds to VEGF, VEGFR1, EGFR, ErbB2, IGF-IR, cMET, FGFR1, FGFR2, and CD20.
  • the antibody of the MRD-containing antibody binds to EGFR.
  • the antibody is Erbitux®, nimotuzumab, or zalutumumab (e.g., Genmab).
  • the antibody binds to the same epitope as Erbitux® antibody or competitively inhibits binding of the Erbitux® antibody to EGFR.
  • the antibody is the Erbitux® antibody.
  • the antibody binds to the same epitope as Erbitux®, nimotuzumab, zalutumumab (e.g., Genmab) antibody.
  • the antibody component, MRD component, and/or MRD-containing antibody competitively inhibits binding of Erbitux®, nimotuzumab, zalutumumab antibody to EGFR.
  • an MRD-containing antibody binds EGFR and a target selected from: HGF, CD64, CDCP1, RON, cMET, ErbB2, ErbB3, IGF1R, PLGF, RGMa, PDGFRa, PDGFRh, VEGFR1, VEGFR2, TNFRSF10A (DR4), TNFRSF10B (DR5), IGF1,2, IGF2, CD3, CD4, NKG2D and tetanus toxoid.
  • the multivalent and monovalent multispecific composition binds at least 1, 2, 3, 4, 5 or more of these targets.
  • the antibody component of the MRD-containing antibody binds EGFR.
  • the antibody component of the MRD-containing antibody is nimotuzumab, zalutumumab.
  • the antibody component of the MRD-containing antibody is Erbitux®.
  • the antibody of the MRD-containing antibody binds to ErbB2.
  • the antibody is HERCEPTIN® (trastuzumab) antibody or competitively inhibits HERCEPTIN® (trastuzumab) antibody binding to ErbB2.
  • the antibody binds to VEGF. In another specific embodiment, the antibody binds to the same epitope as AVASTIN® (bevacizumab) antibody or competitively inhibits AVASTIN® antibody. In a further specific embodiment, the antibody is the AVASTIN® antibody.
  • the antibody binds to a target that is associated with a disease or disorder of the immune system. In one embodiment, the antibody binds to TNF. In another specific embodiment, the antibody binds to the same epitope as HUMIRA® (adalimumab) antibody or competitively inhibits HUMIRA® antibody. In a further specific embodiment, the antibody is the HUMIRA® antibody. In one embodiment, the antibody binds to TNF. In another specific embodiment, the antibody binds to the same epitope as SIMPONITM (golimumab) antibody or competitively inhibits SIMPONITM antibody. In a further specific embodiment, the antibody is the SIMPONITM antibody.
  • the antibody component of the MRD containing antibody binds to a target that is associated with a disease or disorder of the metabolic, cardiovascular, musculoskeletal, neurological, or skeletal system. In other embodiments, the antibody component of the MRD containing antibody binds to a target that is associated with yeast, fungal, viral or bacterial infection or disease.
  • the MRD is about 2 to 150 amino acids. In another embodiment, the MRD is about 2 to 60 amino acids.
  • MRDs can be linked to an antibody or other MRDs directly or through a linker.
  • the MRDs can be any target binding peptide.
  • the MRD target is a soluble factor.
  • the MRD target is a transmembrane protein such as a cell surface receptor.
  • the target of the MRD is a cellular antigen. In a specific embodiment, the target of the MRD is CD20.
  • the target of the MRD is an integrin.
  • the peptide sequence of the integrin targeting MRD is YCRGDCT (SEQ ID NO:3).
  • the peptide sequence of the integrin targeting MRD is PCRGDCL (SEQ ID NO:4).
  • the peptide sequence of the integrin targeting MRD is TCRGDCY (SEQ ID NO:5).
  • the peptide sequence of the integrin targeting MRD is LCRGDCF (SEQ ID NO:5).
  • the target of the MRD is an angiogenic cytokine.
  • the peptide sequence of the angiogenic cytokine targeting (i.e., binding) MRD is MGAQTNFMPMDDLEQRLYEQFILQQGLE (SEQ ID NO:7).
  • the target of the MRD is ErbB2. In another embodiment, the target to which the MRD binds is ErbB3. In an additional embodiment, the target to which the MRD binds is tumor-associated surface antigen or an epithelial cell adhesion molecule (Ep-CAM).
  • Ep-CAM epithelial cell adhesion molecule
  • the target to which the MRD binds is VEGF.
  • the peptide sequence of the VEGF targeting MRD is VEPNCDIHVMWEWECFERL (SEQ ID NO:13).
  • the target to which the MRD binds is an insulin-like growth factor-I receptor (IGF1R).
  • IGF1R targeting MRD includes, for example, a peptide sequence having the formula: NFYQCIDLLMAYPAEKSRGQWQECRTGG (SEQ ID NO:37);
  • the target of the MRD is a tumor antigen.
  • tumor antigen as used herein may be understood as both those antigens (including mutations) exclusively expressed on tumor cells (i.e., tumor-specific antigens) and those antigens expressed on tumor cells and normal cells (e.g., antigens overexpressed on tumor cells).
  • the target of the MRD is an epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • the target of the MRD is an angiogenic factor.
  • the target of the MRD is an angiogenic receptor.
  • the MRD is a vascular homing peptide.
  • the target of the MRD is a nerve growth factor.
  • the antibody and/or MRD binds to EGFR, ErbB2, ErbB3, ErbB4, CD20, insulin-like growth factor-I receptor, or prostate specific membrane antigen.
  • the present invention also relates to an isolated polynucleotide comprising a nucleotide sequence encoding an MRD-containing antibody.
  • a vector comprises a polynucleotide sequence encoding an MRD-containing antibody.
  • the polynucleotide sequence encoding an MRD-containing antibody is operatively linked with a regulatory sequence that controls expression on the polynucleotide.
  • a host cell comprises the polynucleotide sequence encoding an MRD-containing antibody.
  • multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the present invention also relates to methods of designing and making multivalent and multispecific compositions (e.g., MRD-containing antibodies) having a full-length antibody comprising a MRD.
  • the MRD is derived from a phage display library.
  • the MRD is derived from natural ligands.
  • the MRD is derived from yeast display of RNA display technology.
  • the present invention also relates to a method of treating or preventing a disease or disorder in a subject (patient) in need thereof, comprising administering an antibody comprising an MRD to the subject (patient).
  • the disease is cancer.
  • undesired angiogenesis in inhibited.
  • angiogenesis is modulated.
  • tumor growth is inhibited.
  • Certain embodiments provide for methods of treating or preventing a disease, disorder, or injury comprising administering to a patient in need thereof, a therapeutically effective amount of a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) to a patient in need thereof.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • the disease, disorder or injury is cancer.
  • the disease, disorder or injury is a disorder of the immune system.
  • the disorder of the immune system is inflammation.
  • the disorder of the immune system is an autoimmune disease.
  • the disorder of the immune system is selected from the group consisting of: rheumatoid arthritis, Crohn's disease, systemic lupus erythematous, inflammatory bowel disease, psoriasis, diabetes ulcerative colitis, and multiple sclerosis.
  • the disease, disorder or injury is a metabolic disease.
  • the disease, disorder, or injury is an infectious disease.
  • the infectious disease is human immunodeficiency virus (HIV) infection or AIDS, botulism, anthrax, or clostridium difficile .
  • the disease, disorder, or injury is neurological.
  • the neurological disease, disorder or injury is pain.
  • the pain is, acute pain or chronic pain.
  • a method of treatment or prevention comprising administering an additional therapeutic agent along with an antibody comprising an MRD is provided.
  • the methods of treatment or prevention comprise administering an antibody comprising more than one type of MRD.
  • FIG. 1 shows the schematic representation of different designs of multi-specific and multivalent molecules. MRDs are depicted as triangles, circles, diamonds, and squares.
  • FIG. 2A shows a typical peptibody as a C-terminal fusion with the heavy chain of Fc.
  • FIG. 2B shows an MRD containing antibody with a C-terminal MRD fusion with the light chain of the antibody.
  • FIG. 2C shows an MRD containing antibody with an N-terminal MRD fusion with the light chain of the antibody.
  • FIG. 2D shows an MRD containing antibody with unique MRD peptides fused to each terminus of the antibody.
  • FIG. 3 depicts the results of an enzyme linked immunosorbent assay (ELISA) in which integrin and Ang2 were bound by an anti-integrin antibody (JC7U) fused to an Ang2 targeting MRD (2xCon4).
  • ELISA enzyme linked immunosorbent assay
  • FIG. 4 depicts the results of an ELISA in which, integrin and Ang2 were bound by an anti-integrin antibody (JC7U) fused to an Ang2 targeting MRD (2xCon4).
  • JC7U anti-integrin antibody
  • FIG. 5 depicts the results of an ELISA in which an anti-Erb132 antibody was fused to an MRD which targets Ang2.
  • FIG. 6 depicts the results of an ELISA in which an Ang2 targeting MRD was fused to a hepatocyte, growth factor receptor (cMET) binding antibody.
  • FIG. 7 depicts the results of an ELISA in which an integrin targeting MRD was fused to an ErbB2-binding antibody.
  • FIG. 8 depicts the results of an ELISA in which an integrin targeting MRD was fused to a hepatocyte growth factor receptor binding antibody.
  • FIG. 9 depicts the results of an ELISA in which an insulin-like growth factor-I receptor targeting MRD was fused to an ErbB2-binding antibody.
  • FIG. 10 depicts the results of an ELISA in which a VEGF-targeting MRD was fused to an ErbB2-binding antibody.
  • FIG. 11 depicts the results of an ELISA in which an integrin targeting MRD was fused to a catalytic antibody.
  • FIG. 12 depicts the results of an ELISA in which an Ang2-targeting MRD was fused to a catalytic antibody.
  • FIG. 13 depicts the results of an ELISA in which an integrin targeting MRD and an Ang2 targeting MRD were fused to an ErbB2-binding antibody.
  • FIG. 14 depicts the results of an ELISA in which an integrin targeting MRD was fused to an ErbB2-binding antibody.
  • FIG. 15 depicts the results of an ELISA in which an integrin, Ang2, or insulin-like growth factor-I receptor-targeting MRD was fused to an ErbB2 or hepatocyte growth factor receptor-binding antibody with a short linker peptide.
  • FIG. 16 depicts the results of an ELISA in which an integrin, Ang2, or insulin-like growth factor-I receptor-targeting MRD was fused to an ErbB2 or hepatocyte growth factor receptor-binding antibody with a long linker peptide.
  • FIG. 17A depicts the dose response curves of MRD-maltose binding protein (MBP) fusions assayed for direct binding to Ang2.
  • MRP MRD-maltose binding protein
  • FIG. 17B indicates MRD-MBP fasion proteins tested, the amino acid sequence of the MRD, and the EC50 values (calculated using a 4 parameter fit).
  • the MXD sequence motif in the MRD components of the MRD-MBP fusions is underlined and mutated residues are in bold and italics.
  • FIG. 18A depicts the results of an assay for direct binding of a HERCEPTIN® based zybody (i.e. an MRD containing HERCEPTIN® antibody sequences) antibody-MRDs and a HERCEPTIN® antibody to Her2 (ErbB2) Fc in the presence of biotinylated Ang2. Binding was detected with HRP-conjugated anti-human kappa chain mAb.
  • a HERCEPTIN® based zybody i.e. an MRD containing HERCEPTIN® antibody sequences
  • Her2 (ErbB2) Fc Her2
  • FIG. 18B depicts the results of an assay for direct binding of a HERCEPTIN® based zybody (i.e., an MRD containing HERCEPTIN® antibody sequences) and a HERCEPTIN® antibody to Her2 Fc in the presence of biotinylated Ang2. Binding was detected with horseradish peroxidase (HRP)-conjugated streptavidin.
  • HRP horseradish peroxidase
  • FIG. 19A depicts the results of an assay for direct binding of anti body-MRDs and an AVASTIN® antibody to VEGF in the presence of biotinylated Ang2. Binding was detected with HRP-conjugated anti-human kappa chain mAb.
  • FIG. 19B depicts the results of an assay for direct binding of antibody-MRDs and an AVASTIN® antibody to VEGF in the presence of biotinylated Ang2. Binding was detected with HRP-conjugated streptavidin.
  • FIG. 20A depicts the results of a flow cytometry assay which demonstrates that antibody-MRDs simultaneously bind Her2 and Ang2 on BT-474 breast cancer cells.
  • FIG. 20B depicts binding of antibody-MRDs to HER2 on BT-474 breast cancer cells.
  • FIG. 21 depicts the results of an ELISA assay that demonstrates the inhibitory effect of antibody-MRDs on TIE-2 binding to plate immobilized Ang2.
  • FIG. 22 depicts the results of a competitive binding assay that demonstrates the inhibition of binding of biotinylated antibody by antibody-MRD and unlabeled antibody.
  • FIG. 23 depicts the results of a competitive binding assay that illustrates the inhibition of labeled antibody binding to BT-474 cells by antibody-MRDs and unlabeled antibody.
  • FIG. 24A depicts the fitted dose curves illustrating the inhibition of BT-474 cell proliferation by HERCEPTIN® with the lm32 MRD (SEQ ID NO:8) fused to the heavy chain and HERCEPTIN®.
  • FIG. 24B depicts the fitted dose curves illustrating the inhibition of BT-474 cell proliferation by HERCEPTIN® with the lm32 MRD fused to the light chain and HERCEPTIN®.
  • FIG. 24C depicts the fitted dose curves illustrating the inhibition of BT-474 cell proliferation by HERCEPTIN® with the 2xcon4 MRD fused to the heavy chain and HERCEPTIN®.
  • FIG. 25A depicts the results of a cytotoxicity assay illustrating ADCC-mediated killing of BT-474 cells by HERCEPTIN® with the Lm32 MRD fused to the heavy chain, HERCEPTIN® with the lm32 MRD fused to the light chain, and HERCEPTIN®.
  • FIG. 25B depicts the results of a cytotoxicity assay illustrating ADCC-mediated killing of BT-474 cells by HERCEPTIN® with the 2xcon4 MRD fused to the heavy chain, and HERCEPTIN®.
  • FIG. 26A depicts the inhibition of HUVEC proliferation by AVASTIN® with the lm32 MRD fused to the heavy chain and AVASTIN® using HUVECs obtained from GlycoTech (Gaithersburg, Md.).
  • FIG. 26B depicts the inhibition of HUVEC proliferation by AVASTIN® with the lm32 MRD fused to the heavy chain and AVASTIN® using HUVECs obtained from Lonza.
  • FIG. 27 depicts the effect of RITUXIMAB®, HERCEPTIN®, and an MRD-containing antibody on tumor volume in vivo.
  • FIG. 28 depicts the increased effect of an antibody-containing MRD on receptor phosphorylation and AKT activation compared to the effect of an antibody in combination with the MRD.
  • FIG. 29A depicts the increased effect of a bispecific MRD-containing antibody on cell proliferation compared to the effect of the antibody or the antibody in combination with the MRD.
  • FIG. 29B depicts the increased effect of a pentaspecific MRD-containing antibody on cell proliferation compared to the effect of the antibody or the antibody in combination with the MRD.
  • FIG. 30 depicts the increased efficacy of a HUMIRA antibody containing an Ang2-binding MRD in an arthritis model compared to HUMIRA.
  • FIG. 31 shows inhibition of EGF-induced signaling in SK-BR3 cells by zybodies.
  • FIG. 32 shows inhibition of Heregulin-induced signaling in SK-BR3 zybodies.
  • FIG. 33 shows inhibition of EGF and Heregulin-induced signaling in SK-BR3 cells by zybodies.
  • FIG. 34 shows a bar-graph (A) and flow-cytometry results (B) depicting the down-regulation of EGFR expression on SK-BR3 cells by zybodies.
  • FIG. 35 shows down-regulation of EGFR in SKBR3 cells by zybodies.
  • the following provides a description of antibodies containing at least one modular recognition domain (MRD).
  • MRD modular recognition domain
  • the linkage of one or more MRDs to an antibody results in a multi specific molecule of the invention that retains structural and functional properties of traditional antibodies or Fc optimized antibodies and can readily be synthesized using conventional antibody expression systems and techniques.
  • the antibody can be any suitable antigen-binding immunoglobulin, and the MRDs can be, any suitable target-binding peptide.
  • the MRDs can be operably linked to any location on the antibody, and the attachment can be direct or indirect (e.g., through a chemical or polypeptide linker).
  • Compositions of antibodies comprising an MRD, methods of manufacturing antibodies comprising an MRD, and methods of using antibodies comprising MRDs are also described in the sections below.
  • Standard techniques may be used for recombinant DNA molecule, protein, and antibody production, as well as for tissue culture and cell transformation. Enzymatic reactions and purification techniques are typically performed according to the manufacturer's specifications or as commonly accomplished in the art using conventional procedures such as those set forth in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988) and Sambrook et al., (Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)) (both herein incorporated by reference), or as described herein.
  • Multivalent and monovalent multispecific complexes can contain MRDs, antibodies, cytoxic agents, and binding motifs in addition to MRDs that bind to one or more targets.
  • a multivalent and monovalent multispecific complex e.g., an MRD-containing antibody
  • a cytotoxic agent e.g., a therapeutic agent
  • multivalent and monovalent multispecific complex(es) and “multivalent and monovalent multispecific complexes” as used herein therefore refer to compositions that are able to bind 2 or more targets and that contain one binding site and/or multiple binding sites for different epitopes. Thus, this term is intended to include complexes containing multiple binding sites for each different epitope bound by the complex, or alternatively, complexes that contain at least one single binding site for a different epitope. The different epitopes can be on the same or different targets. Multivalent and monovalent multispecific complexes can be multivalent and, multispecific and can therefore bind two or more targets and have two or more binding sites for each of the targets bound by the complex.
  • Multivalent and monovalent multispecific complexes can also have one (or more) single binding sites for one (or more) target(s) and multiple binding sites for other targets and accordingly, these complexes are monovalent (with respect to the single binding site(s)), multivalent and multispecific. Moreover, multivalent and monovalent multispecific complexes can be monovalent and multispecific and thus, only contain single binding sites for two or more different targets.
  • multivalent and monovalent multispecific complex-drug complex or “MRD-containing antibody-cytotoxic agent” as used herein, refers to a multivalent and monovalent multispecific complex containing one or more cytotoxic agents.
  • cytotoxic agent includes any agent that is detrimental to cells including for example, substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • the term is intended to include a chemotherapeutic agent, a drug moiety (e.g., a cytokine or prodrug), an antibiotic, a radioactive isotope, a chelating ligand (e.g., DOTA, DOTP, DOTMA, DTPA and TETA), a nucleolytic enzyme, a toxins such as a small molecule toxin or enzymatically active toxin of bacterial, fungal, plant or animal origin, including fragments and/or variants of these toxins.
  • a drug moiety e.g., a cytokine or prodrug
  • an antibiotic e.g., an antibiotic, a radioactive isotope, a chelating ligand (e.g., DOTA, DOTP, DOTMA, DTPA and TETA), a nucle
  • the cytotoxic agent is a member selected from auristatin, dolostantin, MMAE, MMAF, a maytansinoid derivative (e.g., the DM1 (N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine), DM3 (N(2′)-deacetyl-N2-(4-mercapto-1-oxopentyl)-maytansine) and DM4 (N(2′)-deacetyl-N2-(4-mercapto-4-methyl-1-oxopentyl)-maytansine).
  • DM1 N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine
  • DM3 N(2′)-deacetyl-N2-(4-mercapto-1-oxopentyl)-maytansine
  • an antibody is used herein to refer to immunoglobulin molecules that are able to bind antigens through an antigen binding domain (i.e., antibody combining site).
  • the term “antibody” includes polyclonal, oligoclonal (mixtures of antibodies), and monoclonal antibodies, chimeric, single chain, and humanized antibodies.
  • the term “antibody” also includes human antibodies.
  • an antibody comprises at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains; CH1, CH2, and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxyl-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the antibody is a homomeric heavy chain antibody (e.g., camelid antibodies) which lacks the first constant region domain (CH1) but retains an otherwise intact heavy chain and is able to bind antigens through an antigen binding domain.
  • the variable regions of the heavy and light chains in the antibody-MRD fusions of the invention contain a functional binding domain that interacts with an antigen.
  • the term “monoclonal antibody” typically refers to a population of antibody molecules that contain only one species of antibody combining site capable of immunoreacting with a particular epitope. A monoclonal antibody thus typically displays a single binding affinity for any epitope with which it immunoreacts.
  • a “monoclonal antibody” may also contain an antibody molecule having a plurality of antibody combining sites (i.e., a plurality of variable domains), each immunospecific for a different epitope, e.g., a bispecific monoclonal antibody.
  • a “monoclonal antibody” refers to a homogeneous antibody population involved in the highly specific recognition and binding of one or two (in the case of a bispecific monoclonal antibody) antigenic determinants, or epitopes. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants.
  • the term “monoclonal antibody” refers to such antibodies made in any number of manners including but not limited to by hybridoma, phage selection, recombinant expression, yeast, and transgenic animals.
  • a “dual-specific antibody” is used herein to refer to an immunoglobulin molecule that contains dual-variable-domain immunoglobulins, where the dual-variable-domain can be engineered from any two monoclonal antibodies.
  • chimeric antibodies refers to antibodies wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species.
  • the variable region of both light and heavy chains corresponds to the variable region of antibodies derived from one species of mammals (e.g., mouse, rat, rabbit, etc.) with the desired specificity and/or affinity while the constant regions are homologous to the sequences in antibodies derived from another species (usually human) to avoid eliciting an immune response in that species.
  • humanized antibody refers to forms of non-human (e.g., murine) antibodies that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human (e.g., murine) sequences.
  • humanized antibodies are human immunoglobulins in which residues from the complementarity determining region (CDR) are replaced by residues from the CDR of a non-human species (e.g., mouse, rat, rabbit, hamster) that have the desired specificity and/or affinity (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature 332:323-327 (1988); Verhoeyen et al., Science 239:1534-1536 (1988)).
  • the Fv framework region (FR) residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity and/or affinity.
  • the humanized antibody can be further modified by the substitution of additional residues either in the Fv framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, affinity, and/or capability.
  • the humanized antibody will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDR regions that correspond to the non-human immunoglobulin whereas all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody can also comprise an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in U.S. Pat. No. 5,225,539, U.S. Pat. No.
  • human antibodies include antibodies having the amino acid sequence of a human immunoglobulin or one or more human germlines and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulins and that do not express endogenous immunoglobulins, as described infra and, for example in, U.S. Pat. No. 5,939,598 by Kucherlapati et al., A human antibody may still be considered “human” even if amino acid substitutions are made in the antibody. Examples of methods used to generate human antibodies are described in: Int. Appl. Publ. Nos. WO98/24893, WO92/01047, WO96/34096, and WO96/33735; European Pat. No.
  • an “antibody combining site” is that structural portion of an antibody molecule comprised of heavy and light chain variable and hypervariable regions that specifically binds (immunoreacts with) an antigen.
  • the term “immunoreact” in its various forms means specific binding between an antigenic determinant-containing molecule and a molecule containing an antibody combining site such as a whole antibody molecule or a portion thereof.
  • each antigen binding domain is short, non-contiguous sequences of amino acids that are specifically positioned to form the antigen binding domain as the antibody assumes its three dimensional configuration in an aqueous environment.
  • the remainder of the amino acids in the antigen binding domains referred to as “framework” regions, show less inter-molecular variability.
  • the framework regions largely adopt a ⁇ -sheet conformation and the CDRs form loops which connect, and in some cases form part of, the ⁇ -sheet structure.
  • framework regions act to form a scaffold that provides for positioning the CDRs in correct orientation by inter-chain, non-covalent interactions.
  • the antigen binding domain (i.e., antibody combining site) formed by the positioned CDRs defines a surface complementary to the epitope on the immunoreactive antigen. This complementary surface promotes the non-covalent binding of the antibody to its cognate epitope.
  • the amino acids comprising the CDRs and the framework regions, respectively, can be readily identified for any given heavy or light chain variable region by one of ordinary skill in the art, since they have been precisely defined (see, “Sequences of Proteins of Immunological Interest,” Kabat, E., et al., U.S. Department of Health and Human Services, (1983); and Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987), which are herein incorporated by reference).
  • “Humanized antibody” or “chimeric antibody” includes antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • T lymphocyte T cell
  • T cells T cell population
  • T lymphocyte T cell
  • T cells T cell population
  • T cells may refer to any T cells, including for example, lymphocytes that are phenotypically CD3 + i.e., express CD3 on the cell surface.
  • CD3 is used to refer individually or collectively to a molecule expressed as part of the T cell receptor and having a meaning as typically ascribed to it in the art.
  • CD3 encompasses all known CD3 subunits, for example CD3 delta, CD3 epsilon, CD3 gamma, and CD3 zeta (TCR zeta), as well as CD3 alpha (TCR alpha), and CD3 beta (TCR beta) in individual or independently combined form.
  • peptibody refers to a peptide or polypeptide which comprises less than a complete, intact antibody.
  • a peptibody can be an antibody Fc domain attached to at least one peptide.
  • a peptibody does not include antibody variable regions, an antibody combining site, CH1 domains, or Ig, light chain constant region domains.
  • Naturally occurring when used in connection with biological materials such as a nucleic acid molecules, polypeptides, host cells, and the like refers to those which are found in nature and not modified by a human being.
  • domain refers to a part of a molecule or structure that shares common physical or chemical features, for example hydrophobic, polar, globular, helical domains or properties, e.g., a protein binding domain, a DNA binding domain or an ATP binding domain. Domains can be identified by their homology to conserved structural or functional motifs.
  • a “conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain.
  • Families of amino acid residues having, similar side chains have been defined in the art, including, basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains
  • substitution of a phenylalanine for a tyrosine is a conservative substitution.
  • conservative substitutions in the sequences of the polypeptides and antibodies of the invention do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence to the antigen(s) to which the polypeptide or antibody binds.
  • Methods of identifying nucleotide and amino acid conservative substitutions and non-conservative substitutions which do not eliminate polypeptide or antigen binding are, well-known in the art (see, e.g., Brummell et al., Biochem. 32:1180-1187 (1993); Kobayashi et al., Protein Eng. 12(10):879-884 (1999); and Burks et al., Proc. Natl. Acad. Sci. USA 94:412-417 (1997)).
  • a “modular recognition domain” (MRD) or “target binding peptide” is a molecule, such as a protein, glycoprotein and the like, that can specifically (non-randomly) bind to a target molecule.
  • the amino acid sequence of a MRD can typically tolerate some degree of variability and still retain a degree of capacity to bind the target molecule. Furthermore, changes in the sequence can result in changes in the binding specificity and in the binding constant between a preselected target molecule and the binding site.
  • the MRD is an agonist of the target it binds.
  • An MRD agonist refers to a MRD that in some way increases or enhances the biological activity of the MRD's target protein or has biological activity comparable to a known agonist of the MRD's target protein.
  • the MRD is an antagonist of the target it binds.
  • An MRD antagonist refers to an MRD that blocks or in some way interferes with the biological activity of the MRD's target protein or has biological activity comparable to a known antagonist or inhibitor of the MRD's target protein.
  • Cell surface receptor refers to molecules and complexes of molecules capable of receiving a signal and the transmission of such a signal across the plasma membrane of a cell.
  • An example of a cell surface receptor of the present invention is an activated integrin receptor, for example, an activated ⁇ v ⁇ 3 integrin receptor on a metastatic cell.
  • cell surface receptor also includes a molecule expressed on a cell surface that is capable of being bound by an MRD containing antibody of the invention.
  • a “target binding site” or “target site” is any known, or yet to be defined, amino acid sequence having the ability to selectively bind a preselected agent.
  • exemplary reference target sites are derived from the RGD-dependent integrin ligands, namely fibronectin, fibrinogen, vitronectin, von Willebrand factor and the like, from cellular receptors such as ErbB2, VEGF, vascular homing peptide or angiogenic cytokines, from protein hormones receptors such as insulin-like growth factor-I receptor, epidermal growth factor receptor and the like, and from tumor antigens.
  • epitopes or “antigenic determinant” are used interchangeably herein and refer to that portion of any molecule capable of being recognized and specifically bound by a particular binding agent (e.g., an antibody or an MRD).
  • a particular binding agent e.g., an antibody or an MRD.
  • epitopes can be formed from contiguous amino acids and noncontiguous amino acids and/or other chemically active surface groups of molecules (such as carbohydrates) juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding are typically lost upon protein denaturing.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • an antibody, MRD, antibody-containing MRD, or other molecule is said to “competitivety inhibit” binding of a reference molecule to a given epitope if it binds to that epitope to the extent that it blocks, to some degree, binding of the reference molecule to the epitope.
  • Competitive inhibition may be determined by any method known in the art, for example, competition ELISA assays.
  • an antibody, MRD, antibody-containing MRD, or other molecule may be said to competitively inhibit binding of the reference molecule to a given epitope, for example, by at least 90%, at least 80%, at least 70%, at least 60%, or at least 50%.
  • protein is defined as a biological polymer comprising units derived from amino acids linked via peptide bonds; a protein can be composed of two or more chains.
  • a “fusion polypeptide” is a polypeptide comprised of at least two polypeptides and optionally a linking sequence to operatively link the two polypeptides into one continuous polypeptide.
  • the two polypeptides linked in a fusion polypeptide are typically derived from two independent sources, and therefore a fusion polypeptide comprises two linked polypeptides not normally found linked in nature.
  • the two polypeptides may be operably attached directly by a peptide bond or may be linked indirectly through a linker described herein or otherwise known in the art.
  • operably linked indicates that two molecules are attached so as to each retain functional activity. Two molecules are “operably linked” whether they are attached directly (e.g., a fusion protein) or indirectly (e.g., via a linker).
  • linker refers to a peptide located between the antibody and the MRD or between two MRDs.
  • Linkers can have from about 1 to 20 amino acids, about 2 to 20 amino acids, or about 4 to 15 amino acids. One or more of these amino acids may be glycosylated, as is well understood by those in the art.
  • the 1 to 20 amino acids are selected from glycine, alanine, proline, asparagine, glutamine, and lysine.
  • a linker is made up of a majority of amino acids that are sterically unhindered, such as glycine and alanine.
  • the linker is selected from polyglycines (such as (Gly) 5 , and (Gly) 8 ), poly(Gly-Ala), and polyalanines.
  • the linker can also be a non-peptide linker such as an alkyl linker, or a PEG linker.
  • alkyl linkers may further be substituted by any non-sterically hindering group such as lower alkyl (e.g., C 1 -C 6 ) lower acyl, halogen (e.g., Cl, Br), CN, NH 2 , phenyl, etc.
  • An exemplary non-peptide linker is a PEG linker.
  • the PEG linker has a molecular weight of about 100 to 5000 kDa, or about 100 to 500 kDa.
  • the peptide linkers may be altered to form derivatives.
  • the linker is a non-peptide linker such as an alkyl linker, or a PEG linker.
  • the linker is a “cleavable linker” facilitating release of an MRD or cytotoxic agent within a cell or in the proximity of the cell.
  • Target cell refers to any cell in a subject (e.g., a human or animal) that can be targeted by a multispecific and multivalent composition (e.g., an antibody-containing MRD) or MRD of the invention.
  • the target cell can be a cell expressing or overexpressing the target binding site, such as an activated integrin receptor.
  • immune response refers to the action of, for example, lymphocytes, antigen presenting cells, phagocytic cells, granulocytes, and soluble macromolecules produced by the above cells or the liver (including antibodies, cytokines, and complement) that results in selective damage to, destruction of, or elimination from the human body of invading pathogens, cells, or tissues infected with pathogens, cancerous cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.
  • effector cell refers to an immune cell which is involved in the effector phase of an immune response, as opposed to the cognitive and activation phases of an immune response.
  • exemplary immune cells include a cell of a myeloid or lymphoid origin, e.g., lymphocytes (e.g., B cells and T cells including cytolytic T cells (CTLs)), killer cells, natural killer cells, macrophages, monocytes, eosinophils, neutrophils, polymorphonuclear cells, granulocytes, mast cells, and basophils).
  • lymphocytes e.g., B cells and T cells including cytolytic T cells (CTLs)
  • killer cells e.g., natural killer cells, macrophages, monocytes, eosinophils, neutrophils, polymorphonuclear cells, granulocytes, mast cells, and basophils.
  • an effector cell is capable of inducing antibody-dependent cell-mediated cytotoxicity (ADCC), e.g., a neutrophil capable of inducing ADCC.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • monocytes and macrophages which express FcR, are involved in specific killing of target cells and presenting antigens to other components of the immune system, or binding to cells that present antigens.
  • an effector cell can phagocytose a target antigen or target cell.
  • the expression of a particular FcR on an effector, cell can be regulated by humoral factors such as cytokines.
  • Fc alpha RI has been found to be up-regulated by G-CSF or GM-CSF. This enhanced expression increases the effector function of Fc alpha RI-bearing cells against targets.
  • Exemplary functions of an effector cell include the phagocytosing or lysing of a target antigen, or a target cell.
  • Target cell refers to any cell or pathogen whose elimination would be beneficial in a patient (e.g., a human or animal) and that can be targeted by a composition (e.g., antibody) of the invention.
  • “Patient,” “subject,” “animal” or “mammal” are used interchangeably and refer to mammals such as human patients and non-human primates, as well as experimental animals such as rabbits, rats, and mice, and other animals. Animals include all vertebrates, e.g., mammals and non-mammals, such as sheep, dogs, cows, chickens, amphibians, and reptiles. In some embodiments, the patient is a human.
  • Treating” or “treatment” includes the administration of the antibody comprising an MRD of the present invention to prevent or delay the onset of the symptoms, complications, or biochemical indicia of a disease, condition, or disorder, alleviating the symptoms or arresting or inhibiting further development of the disease, condition, or disorder.
  • Treatment can be prophylactic (to prevent or delay the onset of the disease, or to prevent the manifestation of clinical or subclinical symptoms thereof) or therapeutic suppression or alleviation of symptoms after the manifestation of the disease, condition, or disorder.
  • Treatment can be with the antibody-MRD composition alone, the MRD alone, or in combination of either with one or more additional therapeutic agents.
  • compositions, carriers, diluents and reagents are used interchangeably and represent that the materials are capable of administration to or upon a human without the production of therapeutically prohibitive undesirable physiological effects such as nausea, dizziness, gastric upset and the like.
  • Modulate means adjustment or regulation of amplitude, frequency, degree, or activity.
  • modulation may be positively modulated (e.g., an increase in frequency, degree, or activity) or negatively modulated (e.g., a decrease in frequency, degree, or activity).
  • Cancer “tumor,” or “malignancy” are used as synonymous terms and refer to any of a number of diseases that are characterized by uncontrolled, abnormal proliferation of cells, the ability of affected cells to spread locally or through the bloodstream and lymphatic system to other parts of the body (metastasize) as well as any of a number of characteristic structural and/or molecular features.
  • a “cancerous tumor,” or “malignant cell” is understood as a cell having specific structural properties, lacking differentiation and being capable of invasion and metastasis. Examples of cancers that may be treated using the antibody-MRD fusions of the invention include solid tumors and hematologic cancers.
  • cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancers.
  • Other types of cancer and tumors that may be treated using multivalent and multispecific compositions (e.g., MRD-containing antibodies) are described herein or otherwise known in the art.
  • an “effective amount” of an antibody, MRD, or MRD-containing antibody as disclosed herein is an amount sufficient to carry out a specifically stated purpose such as to bring about an observable change in the level of one or more biological activities related to the target to which the antibody, MRD, or MRD-containing antibody binds.
  • the change increases the level of target activity.
  • the change decreases the level of target activity.
  • An “effective amount” can be determined empirically and in a routine manner, in relation to the stated purpose.
  • therapeutically effective amount refers to an amount of an antibody, MRD, MRD-containing antibody, other multivalent and multispecific drug of the invention, or other drug effective to “treat” a disease or disorder in a patient or mammal.
  • the therapeutically effective amount of the drug can reduce angiogenesis and neovascularization; reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent or stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent or stop) tumor metastasis; inhibit, to some extent, tumor growth or tumor incidence; stimulate immune responses against cancer cells and/or relieve to some extent one or more of the symptoms associated with the cancer. See the definition herein of “treating”.
  • a “therapeutically effective amount” also may refer to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • a therapeutically effective amount of a composition of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the composition to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the therapeutic composition are outweighed by the therapeutically beneficial effects.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, since a prophylactic dose is used in subjects (patients) prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • the present invention encompasses not only the entire group, listed as a whole, but also each member of the group individually and all possible subgroups of the main group, and also the main group absent one or more of the group members.
  • the present invention also envisages the explicit exclusion of one or more of any of the group members in the disclosed and/or claimed invention.
  • the present invention describes an approach based on the adaptation of target binding peptides or modular recognition domains (MRDs) as fusions to catalytic or non-catalytic antibodies.
  • MRDs modular recognition domains
  • the MRD-antibody fusions provide for effective targeting to tumor cells or soluble molecules while leaving the prodrug activation capability of the catalytic antibody intact. MRDs can also emend the binding capacity of non-catalytic antibodies providing for an effective approach to extend the binding functionality of antibodies, particularly for therapeutic purposes.
  • a full-length antibody comprising at least one modular recognition domain (MRD).
  • the full-length antibody comprises more than one MRD, wherein the MRDs have the same or different specificities.
  • a single MRD may be comprised of a tandem repeat of the same or different amino acid sequence that can allow for the binding of a single MRD to multiple targets and/or to a repeating epitope on a given target.
  • the interaction between a protein ligand and its target receptor site often takes place at a relatively large interface. However, only a few key residues at the interface contribute to most of the binding.
  • the MRDs can mimic ligand binding. In certain embodiments, the MRD can mimic the biological activity of a ligand (an agonist MRD) or through competitive binding inhibit the bioactivity of the ligand (an antagonist MRD).
  • MRDs in multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRDs of the present invention will generally contain a peptide sequence that binds to target sites of interests and have a length of about 2 to 150 amino acids, about 2 to 125 amino acids, about 2 to 100 amino acids, about 2 to 90 amino acids, about 2 to 80 amino acids, about 2 to 70 amino acids, about 2 to 60 amino acids, about 2 to 50 amino acids, about 2 to 40 amino acids, about 2 to 30 amino acids, or about 2 to 20 amino acids.
  • the MRDs have a length of about 2 to 60 amino acids. In other embodiments, the MRDs have a length of about 10 to 60 amino acids. In other embodiments, the MRDs have a length of about 10 to 50 amino acids. In additional embodiments, the MRDs have a length of about 10 to 40 amino acids. In additional embodiments, the MRDs have a length of about 10 to 30 amino acids.
  • one or more of the MRD components of the multivalent and multispecific compositions have a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 3 M, 10 ⁇ 3 M, 5 ⁇ 10 ⁇ 4 M, 10 ⁇ 4 M, 5 ⁇ 10 ⁇ 5 M, 10 ⁇ 5 M, 5 ⁇ 10 ⁇ 6 M, 10 ⁇ 6 M, 5 ⁇ 10 ⁇ 7 M, 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M, 5 ⁇ 10 ⁇ 13 M, 10 ⁇ 13 M, 5 ⁇ 10 ⁇ 14 M, 10 ⁇ 14 m 5 ⁇ 10 ⁇ 15 M, or 10 ⁇ 15 M.
  • one or more of the MRD components of the multivalent and multispecific compositions have a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 5 M. In another embodiment, one or more of the MRD components of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) have a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 8 M. In another embodiment, one or more of the MRD components of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) have a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 9 M.
  • one or more of the MRD components of the multivalent and multispecific compositions have a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 10 M. In another embodiment, one or more of the MRD components of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) have a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 11 M. In another embodiment, one or more of the MRD components of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) have a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 12 M.
  • one or more of the MRD components of the multivalent and multispecific compositions bind their targets with an off rate (k off ) of less than 5 ⁇ 10 ⁇ 2 sec ⁇ 1 , 10 ⁇ 2 sec ⁇ 1 , 5 ⁇ 10 ⁇ 3 sec ⁇ 1 , or 10 ⁇ 3 sec ⁇ 1 .
  • one or more of the MRD components of the multivalent and multispecific compositions bind their targets with an off rate (k off ) of less than 5 ⁇ 10 ⁇ 4 sec ⁇ 1 , 10 ⁇ 4 sec ⁇ 1 , 5 ⁇ 10 ⁇ 5 sec ⁇ 1 , or 10 ⁇ 5 sec ⁇ 1 , 5 ⁇ 10 ⁇ 6 sec ⁇ 1 , 10 ⁇ 6 sec ⁇ 1 , 5 ⁇ 10 ⁇ 7 sec ⁇ 1 , or 10 ⁇ 7 sec ⁇ 1 .
  • one or more of the MRD components of the multivalent and multispecific compositions bind their targets with an on rate (k on ) of greater than 10 3 M ⁇ 1 sec ⁇ 1 , 5 ⁇ 10 3 M ⁇ 1 sec ⁇ 1 , 10 4 M ⁇ 1 sec ⁇ 1 , or 5 ⁇ 10 4 M ⁇ 1 sec ⁇ 1 .
  • the MRDs are affibodies.
  • Affibodies represent a class of affinity proteins based on a 58-amino acid residue protein domain derived from one of the IgG-binding domains of staphylococcal protein A. This three helix bundle domain has been used as a scaffold for the construction of combinatorial phagemid libraries, from which affibody variants that bind a desired target molecule, such as one or more of the targets disclosed herein, can routinely be selected using phage display technology (see, e.g., Nord et al., Nat. Biotechnol. 15:772-7 (1997), and Ronmark et al., A, Eur. J. Biochem. 2002; 269:2647-55). Further details of Affibodies and methods of production thereof are provided by reference to U.S. Pat. No. 5,831,012, which is herein incorporated by reference in its entirety.
  • an MRD of the invention contains one or more amino acid residues or sequences of amino acid residues (including derivatives, analogs, and mimetics thereof) that are preferentially targeted by chemistries or other processes that covalently or non-covalently link a molecular entity to the MRD, as compared to, the MRD without the preferentially targeted sequences or the antibody component of the MRD-containing antibody.
  • the amino acid sequence of the MRD contains one or more residues having a reactive side chain (e.g. cysteine or lysine) that allows for selective or preferential linkage of the MRD to cytotoxic agents (e.g., drug and prodrug conjugates, toxins, and bioactive ligands) or imaging agents.
  • the use of these “linking” MRDs to arm an MRD-comprising antibody with a “payload” overcomes many of the issues associated with antibody destabilization and reduction in antibody activity that have frequently been observed using conventional methods for generating immunotoxins.
  • the “payload” component of an MRD-comprising antibody complex of the invention can be any composition that confers a beneficial therapeutic, diagnostic, or prognostic effect, or that provide an advantage in manufacturing, purifying or formulating an MRD-containing antibody.
  • the payload is a chemotherapeutic drug, or a prodrug, such as, doxorubicin or a maytansinoid-like drug.
  • the MRD does not contain an antigen binding domain, or another antibody domain such as a constant region, a variable region, a complementarity determining region (CDR), a framework region, an Fc domain, or a hinge region.
  • the MRD does not contain an antigen binding domain.
  • the MRD does not contain three CDRs.
  • the MRD does not contain CDR1 and CDR2.
  • the MRD does not contain CDR1.
  • the MRD is not derived from a natural cellular ligand.
  • the MRD is not a radioisotope.
  • the MRD is not a protein expression marker such as glutathione S-transferase (GST), His-tag, Flag, hemagglutinin (HA), MYC or a fluorescent protein (e.g., GFP or RFP).
  • GST glutathione S-transferase
  • His-tag His-tag
  • Flag hemagglutinin
  • MYC a fluorescent protein
  • the MRD does not bind serum albumin.
  • the MRD is not a small molecule that is a cytotoxin.
  • the MRD does not have enzymatic activity.
  • the MRD has a therapeutic effect when administered alone and/or when fused to an Fc in a patient or animal model.
  • the MRD has a therapeutic effect when repeatedly administered alone and/or when fused to an Fc in a patient or animal model (e.g., 3 or more times over the course of at least six months).
  • the MRD is conformationally constrained. In other embodiments, the MRD is not conformationally constrained. In some embodiments, the MRD contains one cysteine residue.
  • the cysteine residue in the MRD can form an interchain bond (e.g., between cysteines within the same MRD, different peptide linked MRDs, and an MRD and a peptide linked immunoglobulin).
  • the MRD(s) participating in the interchain bond is/are associated with a single core target-binding domain. In other embodiments, the MRD(s) participating in the interchain bond is/are associated with multiple core target-binding domains.
  • the cysteine residue in the MRD can form an interchain bond (e.g., between cysteines of non-peptide linked MRDs or an MRD and an immunoglobulin that are not linked by a peptide bind).
  • the MRD(s) associated with the interchain bond is/are associated with a single core target-binding domain (i.e., 2 MRDs located on different polypeptide chains form one or more interchain bonds and collectively form one target binding site).
  • the invention encompasses MRD-containing antibodies wherein MRDs located on the carboxyl terminus of the heavy chain interact (e.g., via disulfide bond) so as to form a single target binding site.
  • the MRD(s) associated with the interchain bond is/are associated with multiple core target-binding domains.
  • the MRD can contain one or more cysteine residues (or other residue having a reactive side chain (e.g., lysine)) that allows for selective or preferential linkage of the MRD to a cytotoxic agent.
  • the MRD contains two cysteine residues outside the core target-binding domain. In some embodiments, the MRD contains two cysteine residues located within the core target-binding domain at each end of the target-binding domain. In some embodiments, a first cysteine is located near the terminus of the molecule (i.e. at the C-terminus of an MRD on the C-terminus of a linker or antibody chain or at the N-terminus of an MRD on the N-terminus of a linker or antibody chain). Thus, in some embodiments, a first cysteine is located within one amino acid, within two amino acids, within three amino acids, within four amino acids, within five amino acids, or within six amino acids of the terminus, of the molecule.
  • a second cysteine is located near the MRD fusion location (i.e. at the N-terminus of an MRD on the C-terminus of a linker or antibody chain or at the C-terminus of an MRD on the N-terminus of a linker or antibody chain).
  • a second cysteine is located within one amino acid, within two amino acids, within three amino acids, within four amino acids, within five amino acids, within 10 amino acids, or within 15 amino acids from the MRD fusion.
  • the MRD is capped with stable residues. In some embodiments, the MRD is disulfide capped. In some embodiments, the MRD does not contain cleavage sites.
  • the MRD has been selected to not contain known potential human T-cell epitopes.
  • the MRD has a particular hydrophobicity.
  • the hydrophobicity of MRDs can be compared on the basis of retention times determined using hydrophobic interaction chromatography or reverse phase liquid chromatography.
  • the MRD target can be any molecule that it is desirable for an MRD-containing antibody to interact with.
  • the MRD target can be a soluble factor or a transmembrane protein, such as a cell surface receptor.
  • the MRI) target can also be an extracellular component or an intracellular component.
  • the MRD target is a factor that regulates cell proliferation, differentiation, or survival.
  • the MRD target is a cytokine.
  • the MRD target is a factor that regulates angiogenesis.
  • the MRD target is a factor that regulates cellular adhesion and/or cell-cell interaction.
  • the MRD target is a cell signaling molecule.
  • the MRDs are able to bind their respective target when the MRDs are attached to an antibody. In some embodiments, the MRD is able to bind its target when not attached to an antibody. In some embodiments, the MRD is a target agonist. In other embodiments, the MRD is a target antagonist. In certain embodiments, the MRD can be used to localize an MRD-containing antibody to an area where the MRD target is located.
  • MRD sequences can be derived from natural ligands or known sequences that bind to a specific target binding site.
  • phage display technologies have emerged as a powerful method in identifying peptides which bind to target receptors and ligands.
  • naturally occurring and non-naturally occurring (e.g., random peptide) sequences can be displayed by fusion with coat proteins of filamentous phage.
  • the methods for elucidating binding sites on polypeptides using phage display vectors has been previously described, in particular in WO94/18221, which is herein incorporated by reference.
  • the methods generally involve the use of a filamentous phage (phagemid) surface expression vector system for cloning and expressing polypeptides that bind to the pre-selected target site of interest.
  • the methods of the present invention for preparing MRDs include the use of phage display vectors for their particular advantage of providing a means to screen a very large population of expressed display proteins and thereby locate one or more specific clones that code for a desired target binding reactivity.
  • the ability of the polypeptides encoded by the clones to bind a target and/or alter the biological activity of the target can be determined using or routinely modifying assays and other methodologies described herein or otherwise known in the art.
  • phage display technology can be used to identify and improve the binding properties of MRDs. See, e.g., Scott et al., Science 249:386 (1990); Devlin et al., Science 249:404 (1990); U.S. Pat.
  • peptide phage display libraries natural and/or non-naturally occurring peptide sequences can be displayed by fusion with coat proteins of filamentous phage.
  • the displayed peptides can be affinity-eluted against a target of interest if desired.
  • the retained phage may be enriched by successive rounds of affinity purification and repropagation.
  • the best binding peptides may be sequenced to identify key residues within one or more structurally related families of peptides.
  • Structural analysis of protein-protein interaction may also be used to suggest peptides that mimic the binding activity of large protein ligands.
  • the crystal structure may suggest the identity and relative orientation of critical residues of the large protein ligand, from which a peptide such as an MRD may be designed. See, e.g., Takasaki et al., Nature Biotech. 15:1266-1270 (1997).
  • These analytical methods may also be used to investigate the interaction between a target and an MRD selected by phage display, which can suggest further modification of the MRDs to increase binding affinity.
  • a peptide library can be fused to the carboxyl terminus of the lac repressor and expressed in E. coil .
  • Another E. coli -based method allows display on the cell's outer membrane by fusion with as peptidoglycan-associated lipoprotein (PAL). These and related methods are collectively referred to as “ E. coli display.”
  • PAL peptidoglycan-associated lipoprotein
  • translation of random RNA is halted prior to ribosome release, resulting in a library of polypeptides with their associated RNA still attached. This and related methods are collectively referred to as “ribosome display.”
  • Other known methods employ chemical linkage of peptides to RNA.
  • RNA-peptide screening RNA display and mRNA display.
  • Chemically derived peptide libraries have been developed in which peptides are immobilized on stable, non-biological materials, such as polyethylene rods or solvent-permeable resins.
  • Another chemically derived peptide library uses photolithography to scan peptides immobilized on glass slides. These and related methods are collectively referred to as “chemical-peptide screening.” Chemical-peptide screening may be advantageous in that it allows use of D-amino acids and other unnatural analogues, as well as non-peptide elements.
  • An improved MRD that specifically binds a desired target can also be prepared based on a known MRD sequence. For example, at least one, two, three, four, five, or more amino acid mutations (e.g., conservative or non-conservative substitutions), deletions or insertions can be introduced into a known MRD sequence and the resulting MRD can be screened for binding to the desired target and biological activity, such as the ability to antagonize target biological activity or to agonize target biological activity.
  • the sites selected for modification are affinity matured using phage display techniques known in the art. See, e.g., Lowman, Ann. Rev. Biophys. Biomol. Struct. 26:401-4 24 (1997).
  • Any technique for mutagenesis known in the art can be used to modify individual nucleotides in a DNA sequence, for purposes of making amino acid addition(s), substitution(s) or deletion(s) in the antibody sequence, or for creating/deleting restriction sites and sequences coding for desired amino acids (e.g., cysteine) to facilitate further manipulations.
  • Such techniques include, but are not limited to, chemical mutagenesis, in vitro site-directed mutagenesis (Kunkel, Proc. Natl. Acad. Sci. USA 82:488 (1985); Hutchinson et al., J. Biol. Chem. 253:6551 (1978)), oligonucleotide-directed mutagenesis (Smith, Ann. Rev. Genet.
  • DNA shuffling can be employed to alter the activities of SYNAGIS® or fragments thereof (e.g., an antibody or a fragment thereof with higher affinities and lower dissociation rates). See, generally, U.S. Pat. Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458, and Patten et al, Curr. Opinion Biotechnol. 8:724-33 (1997); Harayama et al., Trends Biotechnol.
  • MRDs can be identified based on their effects in assays that measure particular pathways or activities. For example, assays that measure signaling pathways (e.g., phosphorylation studies or multimerization), ion channel fluxes, intracellular cAMP levels, cellular activities such as migration, adherence, proliferation, or apoptosis, and viral entry, replication, budding, or integration can be used to identify, characterize, and improve MRDs.
  • signaling pathways e.g., phosphorylation studies or multimerization
  • ion channel fluxes e.g., phosphorylation studies or multimerization
  • intracellular cAMP levels e.g., phosphorylation studies or multimerization
  • cellular activities such as migration, adherence, proliferation, or apoptosis
  • viral entry, replication, budding, or integration can be used to identify, characterize, and improve MRDs.
  • variants and derivatives of the MRDs that retain the ability to bind the target antigen are included within the scope of the present invention. Included within variants are insertional, deletional, and substitutional variants, as well as variants that include MRDs presented herein with additional amino acids at the N- and/or C-terminus, including from about 0 to 50, 0 to 40, 0 to 30, 0 to 20 amino acids and the like. It is understood that a particular MRD of the present invention may be modified to contain one, two, or all three types of variants. Insertional and substitutional variants may contain natural amino acids, unconventional amino acids, or both.
  • the MRD contains a sequence with no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 amino acid differences when compared to an MRD sequence described herein.
  • the amino acid differences are substitutions. These substitutions can be conservative or non-conservative in nature and can include unconventional or non-natural amino acids.
  • the MRD contains a sequence that competitively inhibits the ability of an MRD-containing sequence described herein to bind with a target molecule. The ability of an MRD to competitively inhibit another MRD-containing sequence can be determined using techniques known in the art, including ELISA and BIAcore analysis.
  • MRD-target interaction can be assayed as described in the Examples below or alternatively, using in vitro or in vivo binding assays such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, fluorescent immunoassays, protein A immunoassays, and immunohistochemistry (IHC).
  • Assays evaluating the ability of an MRD to functionally affect its target e.g., assays to measure signaling, proliferation, migration etc
  • An improved MRD that has a particular half-life in vivo can also be prepared based on a known MRD sequence. For example, at least one, two, three, four, five, or more amino acid mutations (e.g., conservative or non-conservative substitutions), deletions or insertions can be introduced into a known MRD sequence and the resulting MRD can be screened for increased half-life.
  • variants and derivatives, of the MRDs that retain the ability to bind the target and have an increased half-life can be included in multivalent and multispecific compositions (e.g., MRD-containing antibodies).
  • an MRD in an MRD-containing antibody has a half-life of at least about 5, at least about 10, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, at least about 50, at least about 55, at least about 60, at least about 65, at least about 70, at least about 75, at least about 80, at least about 85, at least about 90, at least about 95, at least about 100, at least about 110, at least about 120, at least about 130, at least about 140, or at least about 150 hours.
  • an MRD in an MRD-containing antibody has a half-life of at least about 5, at least about 10, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, at least about 50, at least about 55, at least about 60, at least about 65, at least about 70, at least, about 75, at least about 80, at least about 85, at least about 90, at least about 95, at least about 100, at least about 110, at least about 120, at least about 130, at least about 140, or at least about 150 hours.
  • the peptides may be prepared by any of the methods known in the art.
  • the MRD peptides can be chemically synthesized and operably attached to the antibody or can be synthesized using recombinant technology.
  • MRDs can be synthesized in solution or on a solid support using known techniques.
  • Various automatic synthesizers are commercially available and can be used in accordance with known protocols. See, for example, Tam et al., J. Am. Chem. Soc.
  • MRDs can be synthesized with covalently attached molecules that are not amino acids but aid in the purification, identification, and/or tracking of an MRD in vitro or in vivo. (e.g., biotin for reacting with avidin or avidin-labeled molecules).
  • the MRD targets an integrin.
  • integrins such as ⁇ v ⁇ 3 and ⁇ v ⁇ 5 as tumor-associated markers has been well documented.
  • a recent study of 25 permanent human cell lines established from advanced ovarian cancer demonstrated that all lines were positive for ⁇ v ⁇ 5 expression and many were positive for ⁇ v ⁇ 3 expression.
  • integrin ⁇ v ⁇ 3 and ⁇ v ⁇ 5 antagonists are in clinical development. These include cyclic RGD peptides and synthetic small molecule RGD mimetics.
  • Two antibody-based integrin antagonists are currently in clinical trials for the treatment of cancer. The first is VITAXIN® (MEDI-522 Abegrein), the humanized form of the murine anti-human ⁇ v ⁇ 3 antibody LM609.
  • VITAXIN® MEDI-522 Abegrein
  • Another antibody in clinical trials is CNT095, a fully human Ab that recognizes ⁇ v integrins.
  • CNT095 a fully human Ab that recognizes ⁇ v integrins.
  • a Phase I study of CNT095 in patients with a variety of solid tumors has shown that it is well tolerated.
  • Cliengitide (EMD 121974), a peptide antagonist of ⁇ v ⁇ 3 and ⁇ v ⁇ 5, has also proven safe in phase I trials. Furthermore, there have been numerous drug targeting and imaging studies based on the use of ligands for these receptors. These preclinical and clinical observations demonstrate the importance of targeting ⁇ v ⁇ 3 and ⁇ v ⁇ 5 and studies involving the use of antibodies in this strategy have consistently reported that targeting through these integrins is safe.
  • integrin-binding MRDs containing one or more RGD tripeptide sequence motifs represent an example of MRDs of the invention.
  • Ligands having the RGD motif as a minimum recognition domain and from which MRDs of the invention can be derived are well known, a partial list of which includes, with the corresponding integrin target in parenthesis fibronectin ( ⁇ 3 ⁇ 1, ⁇ 5 ⁇ 1, ⁇ v ⁇ 1, ⁇ 11b ⁇ 3, ⁇ v ⁇ 3, and ⁇ 3 ⁇ 1) fibrinogen ( ⁇ M ⁇ 2 and ⁇ 11b ⁇ 1) on Willebrand factor ( ⁇ 11b ⁇ 3 and ⁇ v ⁇ 3), and vitronectin ( ⁇ 11b ⁇ 3, ⁇ v ⁇ 3 and ⁇ b ⁇ 5).
  • the RGD containing targeting MRD is a member selected from the group consisting of: YCRGDCT (SEQ ID NO:3); PCRGDCL (SEQ ID NO:4); TCRGDCY (SEQ ID NO:5); and LCRGDCF (SEQ ID NO:6).
  • a MRD that mimics a non-RGD-dependent binding site on an integrin receptor and having the target binding specificity of a high affinity ligand that recognizes the selected integrin is also contemplated in the present invention.
  • MRDs that bind to an integrin receptor and disrupt binding and/or signaling activity of the integrin are also contemplated.
  • the MRD targets an angiogenic molecule
  • Angiogenesis is essential to many physiological and pathological processes.
  • Ang2 has been shown to act as a proangiogenic molecule.
  • Administration of Ang2-selective inhibitors is sufficient to suppress both tumor angiogenesis and corneal angiogenesis. Therefore, Ang2 inhibition alone or in combination with inhibition of other angiogenic factors, such as VEGF, can represent an effective antiangiogenic strategy for treating patients with solid tumors.
  • MRDs useful in the present invention include those that bind to angiogenic receptors, angiogenic factors, and/or Ang2.
  • an MRD of the invention binds Ang2.
  • the TIE2 binding component comprises a fragment of ANG2 that binds TIE2.
  • compositions of the invention bind TIE2 and comprise amino acids 283-449 of the human ANG2 disclosed in NCBI Ref. Seq. No. NP — 001138.1.
  • an MRD and/or -MRD-containing antibody binds Ang2 and contains a sequence selected from the group consisting of: GAQTNFMPMDDLEQRLYEQFILQQGLE (SEQ ID NO:9) (ANGa); LWDDCYFFPNPPHCYNSP (SEQ ID NO:11) (ANGb); LWDDC YSYPNPPHCYNSP (SEQ ID NO:12) (ANGc); LWDDCYSFPNPPPICYNSP (SEQ ID NO: 15) (ANGd); DCAVYPNPPWCYKMEFGK (SEQ ID NO:16) (ANGe); PHEECYFYPNPPFICYT MS (SEQ ID NO:17) (ANGf); and PHEECYSYPNPPHCYTMS (SEQ ID NO:18) (ANGg).
  • GAQTNFMPMDDLEQRLYEQFILQQGLE SEQ ID NO:9
  • LWDDCYFFPNPPHCYNSP SEQ ID NO:11
  • ANGb LWD
  • an MRD and/or -MRD-containing antibody binds Ang2 and contains a sequence selected from the group consisting of GAQTNFMPMDDLEQRLYEQPILQ OGLE (SEQ ID NO:9) (ANGa); LWDDCYFFPNPPHCYNSP (SEQ ID NO:11) (ANGb); LWDDCYSYPNPPHCYNSP (SEQ ID NO:12 (ANGc); LWDDCYSFPNPPHCYNSP (SEQ ID NO:15) (ANGd); DCANTYPNPPWCYKMEFGK (SEQ ID NO:16) (ANGe); PHEECYFYPNPP HCYTMS (SEQ ID NO:17) (ANGf); and PHEECYSYPNPPFICYTMS (SEQ ID NO:18) (ANGg).
  • ANG-2 binding peptides disclosed in U.S. Pat. Nos. 7,309,483, 7,205,275, 7,138,370 7,063,965, 7,063,840, 7,045,302, 7,008,781, 6,825,008, 6,645,484, 6,627,415, 6,455,035, 6,441,137, 6,433,143, 6,265,564, 6,166,185, 5,879,672, 5,814,464, 5,681,714, 5,650,490, 5,643,755 and 5,521,073; and U.S. Appl. Publ. Nos.
  • the MRD targets vascular endothelial growth factor (VEGF).
  • the antibody-MRD fusion comprises an MRD with the sequence ATWLPPP (SEQ ID NO:71), which inhibits VEGF-mediated angiogenesis. Binetruy-Tournaire et al., EMBO J. 19:1525-1533 (2000).
  • an anti-VEGF antibody containing an MRD that targets VEGF is contemplated in the present invention.
  • Anti-VEGF antibodies can be found for example in Presta et al., Cancer Research 57:4593-4599 (1997); and Fuh et al., J. Biol. Chem. 281:10 6625 (2006), each of which is herein incorporated by reference in its entirety.
  • Insulin-like growth factor-I receptor-specific MRDs can also be used in the present invention.
  • Vascular homing-specific MRDs are also contemplated for use in the present invention.
  • a number of studies have characterized the efficacy of linking the vascular homing peptide to other proteins like IL12 or drugs to direct their delivery in live animals.
  • target binding sites are contemplated as being the target of the antibody-MRD fusions of the present invention, including for example, FGFR1, FGFR2, EGFR, ErbB2, ErbB3, ErbB4, CD20, insulin-like growth factor-I receptor, and hepatocyte growth factor receptor.
  • MRDs can be directed towards these target binding sites or the corresponding ligands.
  • the MRD binds to IL6. In one embodiment, the MRD binds to IL6R.
  • the MRD binds to HER2/3.
  • the MRD binds ErbB2.
  • the MRD binds to a human protein. In some embodiments, the MRD binds to both a human protein and its ortholog in mouse, rat, rabbit, or hamster.
  • the antibody in the multivalent and multispecific compositions can be any suitable antigen-binding immunoglobulin.
  • the MRD-containing antibody molecules described herein retain the structural and functional properties of traditional monoclonal antibodies.
  • the antibodies retain their epitope binding properties, but advantageously also incorporate one or more additional target-binding specificities.
  • Antibodies that can be used in the multivalent and multispecific compositions include, but are not limited to, monoclonal, multispecific, human, humanized, primatized, and chimeric antibodies.
  • Immunoglobulin or antibody molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule.
  • the antibodies are IgG1.
  • the antibodies are IgG3.
  • Antibodies that can be used as part of the multivalent and multispecific compositions can be naturally derived or the result of recombinant engineering (e.g., phage display antibodies can include xenomouse and synthetic). The modifications, for example, to enhance half-life or to increase or decrease antibody dependent cellular cytotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) activity.
  • Antibodies can be from or derived from any animal origin including birds and mammals or generated synthetically.
  • the antibodies are human, murine, donkey, rabbit, goat, guinea pig, camel, llama, horse, or chicken antibodies. In specific embodiments, the antibodies are human.
  • the heavy chain portions of one polypeptide chain of a multimer are identical to those on a second polypeptide chain of the multimer.
  • the heavy chain portion-containing monomers of the invention are not identical.
  • each monomer may comprise a different target binding site, forming, for example, a bispecific antibody.
  • Bispecific, bivalent antibodies, and methods of making them are described, for instance in U.S. Pat. Nos. 5,731,168, 5,807,706, 5,821,333, and U.S. Appl. Publ. Nos. 2003/020734 and 2002/0155537; each of which is herein incorporated by reference in its entirety.
  • Bispecific tetravalent antibodies, and methods of making them are described, for instance, in Int. Appl. Publ. Nos. WO02/096948 and WO00/44788, the disclosures of both of which are herein incorporated by reference in its entirety. See generally, Int. Appl. Publ. Nos.
  • the heavy chain portions of the antibody component of the MRD-antibody fusions for use in the methods disclosed herein may be derived from different immunoglobulin molecules.
  • a heavy chain portion of a polypeptide may comprise a CH1 domain derived from an IgG1 molecule and a hinge region derived from an IgG3 molecule.
  • a heavy chain portion can comprise a hinge region derived, in part, from an IgG1 molecule and, in part, from an IgG3 molecule.
  • a heavy chain portion can comprise a chimeric hinge region derived, in part, from an IgG1 molecule and, in part, from an IgG4 molecule.
  • the antigen binding domains of the antibody component of the multivalent and multispecific compositions bind to their target with a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 3 M, 10 ⁇ 3 M, 5 ⁇ 10 ⁇ 4 M, 10 ⁇ 4 M, 5 ⁇ 10 ⁇ 5 M, 10 ⁇ 5 M, 5 ⁇ 10 ⁇ 6 M, 10 ⁇ 6 M, 5 ⁇ 10 ⁇ 7 M, 10 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 10 ⁇ 10 M, 10 ⁇ 10 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M, 5 ⁇ 10 ⁇ 13 M, 10 ⁇ 13 M, 5 ⁇ 10 ⁇ 14 M, 10 ⁇ 14 M, 5 ⁇ 10 ⁇ 15 M, or 10 ⁇ 15 M.
  • a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 3 M, 10 ⁇ 3 M, 5 ⁇ 10 ⁇
  • the antibody component of the multivalent and multispecific compositions have a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 5 M.
  • antigen binding of the antibody component of the multivalent and multispecific compositions has a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 8 M.
  • antigen binding of the antibody component of the multivalent and multispecific compositions has a dissociation constant or Kd of less than less than 5 ⁇ 10 ⁇ 9 M.
  • the antibody component of the multivalent and multispecific compositions have a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 10 M. In another embodiment, the antibody component of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) have a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 11 M. In another embodiment, the antibody component of the multivalent and multispecific compositions (e.g., MRD-containing antibodies) have a dissociation constant or Kd of less than 5 ⁇ 10 ⁇ 12 M.
  • the antibody component of the MRD-containing antibody binds its target with an off rate (k off ) of less than 5 ⁇ 10 ⁇ 2 sec ⁇ 1 , 10 ⁇ 2 sec ⁇ 1 , 5 ⁇ 10 ⁇ 3 sec ⁇ 1 , or 10 ⁇ 3 sec ⁇ 1 .
  • the antibody component of the MRD-containing antibody binds its target with an off rate (k off ) of less than 5 ⁇ 10 ⁇ 4 sec ⁇ 1 , 10 ⁇ 4 sec ⁇ 1 , 5 ⁇ 10 ⁇ 5 sec ⁇ 1 , or 10 ⁇ 5 sec ⁇ 1 , 5 ⁇ 10 ⁇ 6 sec ⁇ 1 , 10 ⁇ 6 sec ⁇ 1 , 5 ⁇ 10 ⁇ 7 sec ⁇ 1 , or 10 ⁇ 7 sec ⁇ 1 .
  • the antibody component of the MRD-containing antibody binds its target with an on rate (k on ) of greater than 10 3 M ⁇ 1 sec ⁇ 1 , 5 ⁇ 10 3 M ⁇ 1 sec ⁇ 1 , 10 4 M ⁇ 1 sec ⁇ 1 , or 5 ⁇ 10 4 M ⁇ 1 sec ⁇ 1 . More preferably, the antibody component of the MRD-containing antibody binds its target with an on rate (k on ) of greater than 10 5 M ⁇ 1 sec ⁇ 1 , 5 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 , 10 6 M ⁇ 1 sec ⁇ 1 , or 5 ⁇ 10 6 M ⁇ 1 sec ⁇ 1 , or 10 7 M ⁇ 1 sec ⁇ 1 .
  • Affinity maturation strategies and chain shuffling strategies e.g., gene-shuffling, motif-shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as “DNA shuffling”) are known in the art and can be employed to generate high affinity and/or to alter the activities (e.g., ADCC and CDC) of multivalent and multispecific compositions (e.g., multivalent and multispecific compositions (e.g., MRD-containing antibodies)). See, e.g., U.S. Pat. Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252 and 5,837,458; and Patten et al., Curr.
  • affinity maturation strategies and chain shuffling strategies can routinely be applied to generate multivalent and multispecific compositions (e.g., MRD-containing antibodies) can also include variants and derivatives that improve antibody function and/or desirable pharmacodynamic properties.
  • certain embodiments of the invention include an antibody-MRD fusion, in which at least a fraction of one or more of the constant region domains has been altered so as to provide desired biochemical characteristics such as reduced or increased effector functions, the ability to non-covalently dimerize, increased ability to localize at the site of a tumor, reduced serum half-life, or increased serum half-life when compared with an unaltered antibody of approximately the same immunoreactivity.
  • the alterations of the constant region domains can be amino acid substitutions, insertions, or deletions.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs Fc receptors
  • cytotoxic cells e.g., Natural Killer (NK) cells, neutophils, and macrophages
  • NK Natural Killer
  • Specific high-affinity IgG antibodies directed to the surface of target cells “arm” the cytotoxic cells and are required for such killing. Lysis of the target cell is extracellular, requires contact or close proximity between the cytotoxic cells and target cells, and does not involve complement.
  • the term “enhances ADCC” (e.g., referring to cells) is intended to include any measurable increase in cell lysis when contacted with a variant MRD-containing antibody as compared to the cell killing of the same cell in contact with a MRD-containing antibody that has not been so modified in a way that alters ADCC in the presence of effector cells (for example, at a ratio of target cells:effector cells of 1:50), e.g., an increase in cell lysis by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, or 325%.
  • the antibody component of the antibody-MRD fusion has been modified to increase antibody dependent cellular cytotoxicity (ADCC) (see, e.g., Bruhns et al., Blood 113:3716-3725 (2009); Shields et al., J. Biol. Chem. 276:6591-6604 (2001); Lazar et al., Proc. Natl. Acad. Sci. USA 103:4005-4010 (2006); Stavenhagen et al., Cancer Res., 67:8882-8890 (2007); Horton et al., Cancer Res.
  • ADCC antibody dependent cellular cytotoxicity
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that increases ADCC include one or more modifications corresponding to: IgG1-S298A, E333A, K334A; IgG1-S239D, 1332E; IgG1-S239D, A330L, I332E; IgG1-P247I, A339D or Q; IgG1-D280H, K290S with or without S298D or V; IgG1-F243L, R292P, Y300L; IgG1-F243L, R292P, Y300L, P396L; and IgG1-F243L, R292P, Y300L, V305I, P396L; wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • an Fc variant protein has enhanced ADCC activity relative to a comparable molecule.
  • an Fc variant protein has ADCC activity that is at least 2 fold, or at least 3 fold, or at least 5 fold or at least 10 fold or at least 50 fold or at least 100 fold greater than that of a comparable molecule.
  • an Fc variant protein has enhanced binding to the Fc receptor Fc gamma RIIIA and has enhanced ADCC activity relative to a comparable molecule.
  • the Fc variant protein has both enhanced ADCC activity and enhanced serum half-life relative to a comparable molecule.
  • any particular Fc variant protein to mediate lysis of the target cell by ADCC can be assayed using techniques known in the art.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • immune effector cells which can be activated by the antigen antibody complexes resulting in cytolysis of the target cell. Cytolysis is generally detected by the release of label (e.g., radioactive substrates, fluorescent dyes or natural intracellular proteins) from the lysed cells.
  • label e.g., radioactive substrates, fluorescent dyes or natural intracellular proteins
  • useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • ADCC activity of the multivalent and monovalent multispecific composition can be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes et al., PNAS USA 95:652-656 (1998), and U.S. Pat. No. 7,662,925.
  • the antibody component of the antibody-MRD fusion has been modified to decrease ADCC (see, e.g., Idusogie et al., J. Immunol. 166:2571-2575 (2001); Sazinsky et al., Proc. Natl. Acad. Sci. USA 105:20167-20172 (2008); Davis et al., J. Rheumatol. 34:2204-2210 (2007); Bolt et al., Eur. J. Immunol. 23:403-411 (1993); Alegre et al., Transplantation 57:1537-1543 (1994); Xu et al., Cell Immunol.
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that decreases ADCC include one or more modifications corresponding to: IgG1-K326W, E333S; IgG2-E333S; IgG1-N297A; IgG1-L234A, L235A; IgG2-V234A, G237A; IgG4-L235A, G237A, E318A; IgG4-S228P, L236E; IgG2-EU sequence 118-260; IgG4-EU sequence 261-447; IgG2-H268Q, V309L, A330S, A331S; IgG1-C220S, C226S, C229S, P238S; IgG1-C226S, C229S, E233P, L234V, L235A; and I
  • the antibody component of the antibody-MRD fusion has been modified to increase antibody-dependent cell phagocytosis (ADCP); (see, e.g., Shields et al., J. Biol. Chem. 276:6591-6604 (2001); Lazar et al., Proc. Natl. Acad. Sci. USA 103:4005-4010 (2006); Stavenhagen et al., Cancer Res., 67:8882-8890 (2007); Richards et al., Mol. Cancer. Ther. 7:2517-2527 (2008); Horton et al., Cancer Res.
  • ADCP antibody-dependent cell phagocytosis
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that increases ADCP include one or more modifications corresponding to: IgG1-5298A, E333A, K334A; IgG1-S239D, I332E; IgG1-S239D, A330L, 1332E; IgG1-P247I, A339D or Q; IgG1-D280H, K290S with or without S298D or V; IgG1-F243L, R292P, Y300L; IgG1-F243L, R292P, Y300L, P396L; IgG1-F243L, R292P, Y300L, V305I, P396L; IgG1-G236A, S239D, I332E.
  • the antibody component of the antibody-MRD fusion has been modified to decrease ADCP (see, e.g., Sazinsky et al., Proc. Natl. Acad. Sci. USA 105:20167-20172 (2008); Davis et al., J. Rheumatol. 34:2204-2210 (2007); Bolt et al., Eur. J. Immunol. 23:403-411 (1993); Alegre et al., Transplantation 57:1537-1543 (1994); Xu et al., Cell Immunol. 200:16-20 (2000); Cole et al., Transplantation 68:563-571 (1999); Hutchins et al., Proc. Natl.
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that decreases ADCC include one or more modifications corresponding to: IgG1-N297A; IgGL234A, L235A; IgG2-V234A, G237A; IgG4-L235A, G237A, E318A; IgG4-S228P, L236E; IgG2 EU sequence 118-260; IgG4-EU sequence 261-447; IgG2-H268Q, V309L, A330S, A331S; IgG1-C220S, C226S, C229S, P238S; IgG1-C226S, C229S, E233P, L234V, L235A; and IgG1-L234F, L235E, P331S.
  • “Complement dependent cytotoxicity” and “CDC” refer to the lysing of a target cell in the presence of complement.
  • the complement activation pathway is initiated by the binding of the first component of the complement system (C1q) to a molecule, an antibody for example, complexed with a cognate antigen.
  • C1q the first component of the complement system
  • a CDC assay e.g., as described in Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996), can be performed.
  • an Fc variant protein has enhanced CDC activity relative to a comparable molecule.
  • an Fc variant protein has CDC activity that is at least 2 fold, or at least 3 fold, or at least 5 fold, or at least 10 fold, or at least 50 fold, or at least 100 fold greater than that of a comparable molecule.
  • the Fc variant protein has both enhanced CDC activity and enhanced serum half-life relative to a comparable molecule.
  • the antibody component of the antibody-MRD fusions have been modified to increase complement-dependent cytotoxicity (CDC) (see, e.g., (see, e.g., Idusogie et al., J. Immunol. 166:2571-2575 (2001); Strohl, Curr. Op. Biotechnol. 20:685-691 (2009); and Natsume et al., Cancer Res. 68:3863-3872 (2008), each of which is herein incorporated by reference in its entirety).
  • CDC complement-dependent cytotoxicity
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that increases CDC include one or more modifications corresponding to: IgG1-K326A, E333A; and IgG1-K326W, E333S, IgG2-E333S.
  • the present invention provides formulations, wherein the Fc region comprises a non-naturally occurring amino acid residue at one or more positions selected from the group consisting of 234, 235, 236, 239, 240, 241, 243, 244, 245, 247, 252, 254, 256, 262, 263, 264, 265, 266, 267, 269, 296, 297, 298, 299, 313, 325, 326, 327, 328, 329, 330, 332, 333, and 334 as numbered by the EU index as set forth in Kabat.
  • the Fe region can comprise a non-naturally occurring amino acid residue at additional and/or alternative positions known to one skilled in the art (see, e.g., U.S. Pat. Nos.
  • MRD-containing antibodies of the invention contain an Fc variant comprising at least one non naturally occurring amino acid residue selected from the group consisting of 234D, 234E, 234N, 234Q, 234T, 234H, 234Y, 2341, 234V, 2341, 235A, 235D, 235R, 235W, 235P, 235S, 235N, 235Q, 235T, 235H, 235Y, 2351, 235V, 235F, 236E, 239D, 239E, 239N, 239Q, 239F, 239T, 239H, 239Y, 240I, 240A, 240T, 240M, 241W, 241 L, 241Y, 241E, 241 R, 243W, 243L 243Y, 243R, 243Q, 244H, 245A, 247V, 247G, 252Y, 254T, 256E, 262I, 262A.
  • the Fc region can comprise additional and/or alternative non-naturally occurring amino acid residues known to one skilled in the art (see, e.g., U.S. Pat. Nos. 5,624,821, 6,277,375, and 6,737,056; and Int. Appl. Publ. Nos. WO01/58957. WO02/06919, WO04/016750, WO04/029207, WO04/035752 and WO05/040217).
  • the multivalent and monovalent multispecific composition is an antibody-MRD fusions wherein the antibody component has been modified to increase inhibitory binding to Fc gamma RIIb receptor (see, e.g., Chu et al., Mol. Immunol. 45:3926-3933 (2008)).
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that increases binding to inhibitory Fc gamma RIIb receptor is IgG1-S267E, L328F.
  • the antibody component of the antibody-MRD fusions have been modified to decrease CDC (see, e.g., Int. Appl. Publ. Nos. WO1997/11971 and WO2007/106585; U.S. Appl. Publ. No 2007/0148167A1; McEarchern et al., Blood 109:1185-1192 (2007); Hayden-Ledbetter et al., Clin. Cancer 15:2739-2746 (2009); Lazar et al, Proc. Natl. Acad. Sci. USA 103:4005-4010 (2006); Bruckheimer et al., Neoplasia 11:509-517 (2009); Stohl, Curr. Op.
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that decreases CDC include one or more modifications corresponding to: IgG1-S239D, A330L, I332E; IgG2 EU sequence 118-260; IgG4-EU sequence 261-447; IgG2-H268Q, V309L, A330S, A331S; IgG1-C226S, C229S, E233P, L234V, L235A; IgG1-L234F, L235E, P331S; and IgG1-C226S, P230S.
  • the half-life on an IgG is mediated by its pH-dependent binding to the neonatal receptor FcRn.
  • the antibody component of the antibody-MRD fusion has been modified to enhance binding to FcRn (see, e.g., Petkova et al., Int. Immunol. 18:1759-1769 (2006); Dall'Acqua et al., J. Immunol. 169:5171-5180 (2002); Oganesyan et al., Mol. Immunol. 46:1750-1755 (2009); Dall'Acqua et al., J. Biol. Chem. 281:23514-23524 (2006), Hinton et al., J. Immunol.
  • the antibody of the antibody-MRD fusion has been modified to selectively bind FcRn at pH6.0, but not pH 7.4.
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that increases half-life include one or more modifications corresponding to: IgG1-M252Y, S254T, T256E; IgG1-T250Q, M428L; IgG1-H433K, N434Y; IgG1-N434A; and IgG1-T307A, E380A, N434A.
  • the antibody component of the antibody-MRD fusion has been modified to decrease binding to FcRn (see, e.g., Petkova et al., Int. Immunol. 18:1759-1769 (2006); Datta-Mannan et al., Drug Metab. Dispos. 35:86-94 (2007); Datta-Mannan et al., J. Biol. Chem. 282:1709-1717 (2007); Strohl, Curr. Op. Biotechnol. 20:685-691 (2009); and Vaccaro et al., Nat. Biotechnol. 23:1283-1288 (2005), each of which is herein incorporated by reference in its entirety).
  • Fc sequence engineering modifications contained in the antibody component of the antibody-MRD fusions that decrease half-life include one or more modifications corresponding to: IgG1-M252Y, S254T, T256E; H433K, N434F, 436H; IgG1-1253A; and IgG1-P2571, N434H or D376V, N434H.
  • the antibody-MRD fusions have been glyocoengineered or the Fc portion of the MRD-containing antibody has been mutated to increase effector function using techniques known in the art.
  • the inactivation (through point mutations or other means) of a constant region domain may reduce Fc receptor binding of the circulating modified antibody thereby increasing tumor localization.
  • constant region modifications consistent with the instant invention moderate complement binding and thus reduce the serum half-life and nonspecific association of a conjugated cytotoxin.
  • Yet other modifications of the constant region may be used to modify disulfide linkages or oligosaccharide moieties that allow for enhanced localization due to increased antigen specificity or antibody flexibility.
  • the resulting physiological profile, bioavailability and other biochemical effects of the modifications, such as tumor localization, biodistribution and serum half-life, can easily be measured and quantified using well know immunological techniques without undue experimentation.
  • amino acid substitutions and/or deletions can be generated by mutagenesis methods, including, but not limited to, site-directed mutagenesis (Kunkel, Proc. Natl. Acad. Sci. USA 82:488-492 (1985)), PCR mutagenesis (Higuchi, in “PCR Protocols: A Guide to Methods and Applications”, Academic Press, San Diego, pp. 177-183 (1990)), and cassette mutagenesis (Wells et al., Gene 34:315-323 (1985)).
  • Site-directed mutagenesis can be performed by the overlap-extension PCR method (Higuchi, in “PCR Technology: Principles and Applications for DNA Amplification”, Stockton Press, New York, pp. 61-70 (1989)).
  • the technique of overlap-extension PCR (Higuchi, ibid.) can be used to introduce any desired mutation(s) into a target sequence (the starting DNA).
  • Other methods useful for the generation of antibodies containing non-naturally occurring Fc regions are known in the art (see, e.g., U.S. Pat. Nos.
  • Multivalent and multispecific compositions used according to the methods of the invention also include derivatives that are modified, e.g., by the covalent attachment of any type of molecule to the antibody such that covalent attachment does not prevent the antibody from specifically binding to its cognate, epitope.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, or derivatization by known protecting/blocking groups. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to acetylation, formylation, etc. Additionally, the derivative may contain one or more non-classical amino acids.
  • the antibody component of compositions of the invention is engineered to contain one or more free cysteine amino acids having a thiol reactivity within a desirable range (e.g. 0.6 to 1.0), wherein the cysteine engineered antibody is prepared by a process comprising replacing one or more amino acid residues of a parent antibody by cysteine.
  • a desirable range e.g. 0.6 to 1.0
  • the cysteine engineered antibody is prepared by a process comprising replacing one or more amino acid residues of a parent antibody by cysteine.
  • one or more free cysteine amino acid residues are located in a light chain.
  • one or more free cysteine amino acid residues are located in a heavy chain.
  • one or more free cysteine amino acid residues are located in a both the heavy and light chain.
  • the cysteine engineered MRD-containing antibody contains a free cysteine amino acid having a thiol reactivity value in the range of 0.6 to 1.0, and a sequence modification in the light chain or the heavy chain that is disclosed in U.S. Pat. No. 7,855,275.
  • the cysteine engineered antibody contains a free cysteine amino acid having a thiol reactivity value in the range of 0.6 to 1.0, and a sequence modification in the light chain or the heavy chain that is not disclosed in U.S. Pat. No. 7,855,275, the contents of which are herein incorporated by reference in its entirety.
  • the MRD-containing antibody is engineered to contain one or more free selenocysteine amino acids or another non-natural amino acid capable of forming disulfide bonds.
  • Antibodies containing the same and methods for making such antibodies are known in the art. See, e.g., Hofer et al., Proc. Natl. Acad. Sci. 105(34):12451-12456 (2008); and Hofer et al., Biochem. 48(50):12047-12057 (2009), each of which is herein incorporated by reference in its entirety.
  • one or more free selenocysteine amino acid residues are located in a light chain.
  • one or more free selenocysteine amino acid residues are located in a heavy chain.
  • one or more free selenocysteine amino acid residues are located in a both the heavy and light chain.
  • the multivalent and multispecific compositions have been modified so as to not elicit a deleterious immune response in the animal to be treated, e.g., in a human.
  • the antibody is modified to reduce immunogenicity using art-recognized techniques.
  • antibody components of the multivalent and multispecific compositions e.g., MRD-containing antibodies
  • These types of antibodies are derived from a non-human antibody, typically a murine or primate antibody, that retains or substantially retains the antigen-binding properties of the parent antibody, but which is less immunogenic in humans.
  • CDRs complementarity determining regions
  • De-immunization can also be used to decrease the immunogenicity of an MRD-containing antibody.
  • the term “de-immunization” includes alteration of an MRD-containing antibody to modify T cell epitopes (see, e.g., Int. Appl. Pub. WO9852976A1, and WO0034317A2, each if which is herein incorporated by reference in its entirety).
  • VH and VL sequences from the starting antibody are analyzed and a human T cell epitope “map” is generated from each V region showing the location of epitopes in relation to complementarity-determining regions (CDRs) and other key residues within the sequence.
  • CDRs complementarity-determining regions
  • T cell epitopes from the T cell epitope map are analyzed in order to identify alternative amino acid substitutions with a low risk of altering activity of the final antibody.
  • a range of alternative VH and VL sequences are designed comprising combinations of amino acid substitutions and these sequences are subsequently incorporated into a range of antibodies for use in the diagnostic and treatment methods disclosed herein, which are then tested for function.
  • Typically, between 12 and 24 variant antibodies are generated and tested.
  • Complete heavy and light chain genes comprising modified V and human C regions are then cloned into expression vectors and the subsequent plasmids introduced into cell lines for the production of whole antibody. The antibodies are then compared in appropriate biochemical and biological assays, and the optimal variant is identified.
  • Antibody 38C2 is an antibody-secreting hybridoma and has been previously described in Int. Appl. Pub. WO97/21803. 38C2 contains an antibody combining site that catalyzes the aldol addition reaction between an aliphatic donor and an aldehyde acceptor. In a syngeneic mouse model of neuroblastoma, systemic administration of an etoposide prodrug and intra-tumor injection of Ab 38C2 inhibited tumor growth.
  • the antibody target of the MRD-containing antibody can be any molecule that it is desirable for a MRD-antibody fusion to interact with.
  • the antibody target can be a soluble factor or the antibody target can be a transmembrane protein, such as a cell surface receptor.
  • the antibody target can also be an extracellular component or an intracellular component.
  • the antibody target is a factor that regulates cell proliferation, differentiation, or survival.
  • the antibody target is a cytokine.
  • the antibody target is a factor that regulates angiogenesis.
  • the antibody target is a factor that regulates one or more immune responses, such as, autoimmunity, inflammation and immune responses against cancer cells.
  • the antibody target is a factor that regulates cellular adhesion and/or cell-cell interaction.
  • the antibody target is a cell signaling molecule. The ability of an antibody to bind to a target and to block, increase, or interfere with the biological activity of the antibody target can be determined using or routinely modifying assays, bioassays, and/or animal models known in the art for evaluating such activity.
  • the antibody target of the MRD-containing antibody is a disease-related antigen.
  • the antigen can be an antigen characteristic of a particular cancer, and/or of a particular cell type (e.g., a hyperproliferative cell), and/or of a particular pathogen (e.g., a bacterial cell (e.g., tuberculosis, smallpox, anthrax), a virus (e.g., HIV), a parasite (e.g., malaria, leichmaniasis), a fungal infection, a mold, a mycoplasm , a pr on antigen, or an antigen associated with a disorder of the immune system.
  • a particular pathogen e.g., a bacterial cell (e.g., tuberculosis, smallpox, anthrax), a virus (e.g., HIV), a parasite (e.g., malaria, leichmaniasis), a fungal infection, a mold, a mycoplasm
  • the antibody target of the MRD-containing antibody is a target that has been validated in an animal model or clinical setting.
  • the antibody target of the MRD-containing antibody is a cancer antigen.
  • the antibody target of the MRD-containing antibody is: PDGFRA, PDGFRB, PDGF-A, PDGF-B, PDGF-CC, PDGF-C, PDGF-D, VEGFR1, VEGFR2, VEGFR3, VEGFC, VEGFD, neuropilin 2 (NRP2), betacellulin, PLGF, RET (rearranged during transfection), TIE1, TIE2 (TEK), CA125, CD3, CD4, CD7, CD10, CD13, CD25, CD32, CD32b, CD44, CD49e (integrin alpha 5), CD55, CD64, CD90 (THY1), CD133 (prominin 1), CD147, CD166, CD200, ALDH1, ESA, SHH, DHH, IHH, patched1 (PTCH1), smoothened (SMO), WNT1, WNT2B, WNT3A, WNT4, WNT4A, WNT5A, WNT5B, WNT713, WNT8A, WNT
  • MRD that binds to one of the above targets.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the above antibody and MRD targets and those otherwise described herein are intended to be illustrative and not limiting.
  • the antibody target of the MRD-containing antibody is CD19, CD22, CD30, CD33, CD38, CD44v6, TNFSF5 (CD40 Ligand), TNFRSF5 (CD40), CD52, CD54 (ICAM), CD74, CD80, CD200, EPCAM (EGP2), neuropilin 1 (NRP1), TEM1, mesothelin, TGFbeta 1, TGFBRII, phosphatidlyserine, folate receptor alpha (FOLR1), TNFRSF10A (TRAIL R1 DR4), TNFRSF10B (TRAIL R2 DR5), CXCR4, CCR4, CCL2, HGF, CRYPTO, VLA5, TNFSF9 (41BB Ligand), TNFRSF9 (41BB), CTLA4, HLA-DR, IL6, TNFSF4 (OX40 Ligand), TNFRSF4 (OX40), MUC1, MUC18, mucin CanAg, ganglioside GD3, EG
  • MRD that binds to one of the above targets
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the antibody of the MRD-containing antibody competes for target binding with an antibody selected from: siplizumab CD2 (e.g., MEDI-507, MedImmune), blinatumomab CD19 CD3 (e.g., MT103, Micromet/MedImmune); XMAB®5574 CD19 (Xencor), SGN-19A CD19 (Seattle Genetics), ASG-5ME (Agenesys and Seattle Genetics), MEDI-551 CD19 (MedImmune), epratuzumab CD22 (e.g., hLL2, Immunomedics/UCB), inotuzumab ozogamicin CD22 (Pfizer), iratumumab CD30 (e.g., SGN-30 (Seattle Genetics) and MDX-060 (Medarex)), XMAB®2513 CD30 (Xencor), brentuximab vedotin CD30 (e.g.,
  • XMAB®5485 CD40 Xencor
  • teneliximub ruplizumab CD40L
  • ANTOVA® bivatuzumab mertansine CD44v6, alemtuzumab CD52
  • alemtuzumab CD52 e.g., CAMPATH®/MABCAMPATH®, Genzyme/Bayer
  • BI505 ICAM1 Bioinvent
  • milatuzumab CD74 e.g., antibody of Immunomedics
  • galiximab CD80 Biogen Idec
  • BMS663513 4-1BB Bristol-Myers Squibb
  • Alexion CD200 antibody Alexion
  • edrecolomab EPCAM e.g., MAb17-1A, PANOREX® (GlaxoSmithKline), AT003 EPCAM (Affitech)
  • adecatumumab EPCAM e.g., MT201, Micromet
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the antibody of the MRD-containing antibody competes for target binding with an antibody selected from: MDX-1342 CD19 (BMS), SGN-CD19A CD19 (Seattle Genetics), an anti-CD20 antibody described in U.S. Pat. No. 5,500,362, ofatumumab CD20 (e.g., ARZERRA®, GENMAB), veltuzumab CD20 (hA20, Takeda and Nycomed), PRO70769 CD20 (Genentech; see e.g., Intl. Appl. No.
  • WO2007124299 Novartis
  • IDEC-131 CD40L Biogen
  • MDX-1411 CD70 BMS
  • SGN-75 CD70 ADC SGN-75 CD70 ADC
  • HuMax-CD74TM CD74 ADC Genmab
  • IDEC-114 CD80 Biogen
  • TRC105 CD105/endoglin Tracon
  • ABX-CBL CD147 Amgen
  • AMG888 HER3 Amgen and Daiichi Sankyo
  • HuMV833 VEGF Tsukuba Research Lab, see, e.g., Intl.
  • IMC-11F8 EGFR (Imclone), CDX-110 EGFRvIII (AVANT Immunotherapeutics), zalumumab EGFR (Genmab), 425, EMD55900 and EMD62000 EGFR (Merck KGaA, see, e.g., U.S. Pat. No. 5,558,864), ICR62EGFR (Institute of Cancer Research, see, e.g., Intl. Appl. Publ. No.
  • MRDs that compete for target binding with one of the above antibodies are encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • one of the above-described antibodies is the antibody of the MRD-containing antibody.
  • the antibody of the MRD-containing antibody is an antibody selected from siplizumab CD2 (e.g., MEDI-507, MedImmune), blinatumomab CD19 CD3 (e.g., MT103, Micromet/MedImmune); XMAB®5574 CD19, (Xencor), SGN-19A CD19 (Seattle Genetics), ASG-5ME (Agenesys and Seattle Genetics), MEDI-551 CD19 (MedImmune), epratuzumab CD22 (e.g., hLL2, Immunomedics/UCB), inotuzumab ozogamicin CD22, iratumumab CD30 (e.g., SGN-30 (Seattle Genetics) and MDX-060 (Medarex)), XMAB®2513 CD30 (Xencor), brentuximab vedotin CD30 (e.g., SGN-35, Seattle Genetics),
  • the antibody target of the MRD-containing antibody is ALK1.
  • the antibody is PF-3,446,962 (Pfizer).
  • the antibody binds to the same epitope as PF-3,446,962.
  • the antibody competitively inhibits binding, of PF-3,446,962 to ALK1.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for ALK1 binding with PF-3,446,962 are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is CD22.
  • the antibody is inotuzumab (e.g., inotuzumab ozogamicin CMC-544, PF-5,208,773; Pfizer).
  • the antibody binds to the same epitope as inotuzumab.
  • the antibody competitively inhibits binding of inotuzumab to CD22.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CD22 binding with inotuzumab are also encompassed by the invention.
  • the antibody target of the MRD containing antibody is CRYPTO.
  • the antibody is the Biogen CRYPTO antibody that has advanced to phase I clinical trials (Biogen Idec).
  • the antibody binds to the same epitope as, the Biogen CRYPTO antibody.
  • the antibody competitively inhibits binding of the Biogen CRYPTO antibody to CRYPTO.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CRYPTO binding with the Biogen CRYPTO antibody are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is TNFSF5 (CD40 LIGAND).
  • the antibody is the Biogen CD40L antibody that has advanced to phase I clinical trials (Biogen Idec).
  • the antibody binds to the same epitope as the Biogen CD40L antibody.
  • the antibody competitively inhibits binding of the Biogen CD40L antibody to CD40L.
  • Multivalent and multispecific con positions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CD40L binding with the Biogen CD40L antibody are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is CD80.
  • the antibody is galiximab (Biogen Idec).
  • the antibody binds to the same epitope as galiximab.
  • the antibody competitively inhibits binding of galiximab to CD80.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CD80 binding with galiximab are also encompassed by the invention.
  • an MRD-containing antibody binds CD80 and a target selected from: CD2, CD3, CD4, CD19, CD20, CD22, CD23, CD30, CD33, TNFRSF5 (CD40), CD52, CD74, TNFRSF10A (DR4), TNFRSF10B (DR5), VEGFR1, VEGFR2 and VEGF.
  • an MRD-containing antibody binds CD80 and a target selected from: CD3, CD4 and NKG2D.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind CD80 and also at least bind 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD80.
  • the antibody component of the MRD-containing antibody is galiximab.
  • the antibody target of the MRD-containing antibody is MCSF.
  • the antibody is PD-360,324 (Pfizer).
  • the antibody binds to the same epitope as PD-360,324.
  • the antibody competitively inhibits binding PD-360,324 to MCSF.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for MCSF binding with PD-360,324 are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is CD44.
  • the antibody is PF-3,475,952 (Pfizer).
  • the antibody binds to the same epitope as PF-3,475,952.
  • the antibody competitively inhibits binding of PF-3,475,952 to CD44.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CD44 binding with PF-3,475,952 are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is p-cadherin (CDH3).
  • the antibody is PF-3,732,010 (Pfizer).
  • the antibody binds to the same epitope as PF-3,732,010.
  • the antibody competitively inhibits binding of PF-3,732,010 to p-cadherin.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for p-cadherin binding with PF-3,732,010 are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is ANG2 (ANGPT2).
  • the antibody is MEDI3617 (MedImmune).
  • the antibody binds to the same epitope as MEDI3617.
  • the antibody competitively inhibits binding of MEDI3617 to ANG2.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for ANG2 binding with MEDI3617 are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody is an ANG-2 binding antibody disclosed in U.S. Pat. Nos. 7,063,965, 7,063,840, 6,645,484, 6,627,415, 6,455,035, 6,433,143, 6,376,653, 6,166,185, 5,879,672, 5,814,464, 5,650,490, 5,643,755, 5,521,073; U.S. Appl. Publ. Nos.
  • an MRD-containing antibody binds ANG2 and additionally binds a target selected from: VEGF (i.e., VEGFA), VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNF, FGFR3, EFNa1, EFNa2, ANG1, ANG2, IL1, IL1beta, IL6, IL8, IL18, HGF, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR2, VEGFR3, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ v ⁇ 3
  • Multivalent and multispecific compositions that bind ANG2 and at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody is MEDI3617, AMG780 or REGN910.
  • the antibody component of the MRD-containing antibody is H4L4.
  • the MRD-containing antibody binds ANG2 and TNF. In additional embodiments, the MRD-containing antibody binds ANG2 and IL6. In other embodiments, the MRD-containing antibody binds ANG2 and IL1. In further embodiments, the administered MRD-containing antibody binds ANG2, IL6 and TNF. In further embodiments, the administered MRD-containing antibody binds ANG2, IL1 and TNF. In further embodiments, the MRD-containing antibody binds ANG2, IL1, IL6 and TNF.
  • the MRD-containing antibody binds ANG2 and TNF and the antibody component of the MRD-containing antibody is adalimumab. In another embodiment, the MRD-containing antibody competes with adalimumab for binding to TNF.
  • the antibody component of the MRD-containing antibody binds ANG2.
  • the antibody component of the MRD-containing antibody is an ANG2 binding antibody selected from SAITAng-2-1, SAITAng-2-2, SAITAng-2-3, SAITAng-2-4 or another antibody disclosed in Intl. Appl. Publ. No. WO2009/142460.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having an antibody and/or 1, 2, 3, 4, 5, 6, or more MRDs that compete for ANG2 binding with one or more of these antibodies are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TIE2.
  • the antibody component of the MRD-containing antibody is a TIE2 binding antibody disclosed in U.S. Pat. Nos. 6,365,154 and 6,376,653; U.S. Appl. Publ. Nos. 2007/0025993, 2006/0057138 and 2006/0024297; or Intl. Appl. Publ. Nos. WO2006/020706, WO2000/018437 and WO2000/018804 (the disclosure of each of which is herein incorporated by reference in its entirety).
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having an antibody and/or 1, 2, 3, 4, 5, 6, or more MRDs that compete for TIE binding with one or more these antibodies are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is EGFR(ErbB1), ErbB2, ErbB3, ErbB4, CD20, insulin-like growth factor-I receptor, prostate specific membrane antigen, an integrin, or cMet.
  • the antibody in the MRD-containing antibody specifically binds EGFR(ErbB1).
  • the antibody is ERBITUX® (IMC-C225).
  • the antibody binds to the same epitope as ERBITUX®.
  • the antibody competitively inhibits binding of ERBITUX® to EGFR.
  • the antibody in the MRD-containing antibody inhibits EGFR dimerization.
  • the antibody is matuzumab (e.g., EMD 72000, Merck Serono) or panitumumab (e.g., VECTIBIX®, Amgen).
  • the antibody binds to the same epitope as matuzumab or panitumumab. In another embodiment, the antibody competitively inhibits binding of matuzumab or panitumumab to EGFR. In another embodiment, the antibody is ABX-EGF (Immunex) or MEDX-214 (Medarex). In another embodiment, the antibody binds to the same epitope as ABX-EGF or MEDX-214. In another embodiment, the antibody competitively inhibits binding of ABX-EGF or MEDX-214 to EGFR. In another specific embodiment, the antibody is zalutumumab (Genmab) or nimotazumab (Biocon).
  • the antibody binds to the same epitope as zalutumumab (Genmab) or nimotuzumab (Biocon). In another embodiment, the antibody competitively inhibits binding of zalutumumab (Genmab) or nimotuzumab (Biocon) to EGFR.
  • an MRD-containing antibody binds EGFR(ErbB1) and a target selected from: HGF, CD64, CDCP1, RON, cMET, ErbB2, ErbB3, IGF1R, PLGF, RGMa, PDGFRa, PDGFRb, VEGFR1, VEGFR2, TNFRSF10A (DR4), TNFRSF10B (DR5), IGF1,2, IGF2, CD3, CD4, NKG2D and tetanus toxoid.
  • the multivalent and monovalent multispecific composition binds at least 1, 2, 3, 4, 5 or more of these targets.
  • the antibody component of the MRD-containing antibody binds EGFR.
  • the antibody component of the MRD-containing antibody is matuzumab, panitumumab, MEDX-214, or ABX-EGF.
  • the antibody component of the MRD-containing antibody is nimotuzumab (Biocon) or zalutumumab.
  • the antibody component of the MRD-containing antibody is Erbitux®.
  • the MRD containing antibody binds ErbB1 and additionally binds ErbB3.
  • the antibody component of the MRD-containing antibody binds ErbB1 and an MRD of the MRD-containing antibody binds ErbB3.
  • the antibody component of the MRD-containing antibody is cetuximab.
  • the antibody component of the MRD-containing antibody competes for ErbB1-binding with cetuximab.
  • the antibody in the MRD-containing antibody is an ErbB1-binding antibody selected from: nimotuzumab (Biocon), matuzumab (Merck KGaA), panitumumab (Amgen), zalutumumab (Genmab), MEDX-214, and ABX-EGF.
  • the antibody component, MRD component and/or MRD-containing antibody competes for ErbB1-binding with an antibody selected from nimotuzumab, matuzumab, panitumumab, and zalutumumab.
  • the antibody component of the MRD-containing antibody binds ErbB3 and an MRD of the MRD-containing antibody binds ErbB1
  • the antibody component of the MRD-containing antibody is an ErbB3-binding antibody selected from MM121 (Merrimack), 8B8 (Genentech), AV203 (Aveo), and AMG888 (Amgen).
  • the antibody component, MRD component and/or MRD-containing antibody competes for ErbB3 binding with an antibody selected from MM121, 8B8, AV203, and AMG888.
  • the MRD-containing antibody specifically binds ErbB2 (Her2).
  • the antibody is trastuzumab (e.g., HERCEPTIN®, Genentech/Roche).
  • the antibody binds to the same epitope as trastuzumab.
  • the antibody competitively inhibits binding of trastuzumab to ErbB2.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with at least 1, 2, 3, 4, 5, 6 of the above antibodies.
  • the antibody in the MRD-containing antibody specifically binds to ErbB2.
  • the antibody in the MRD-containing antibody is an antibody that specifically binds to the same epitope as the anti-ErbB2 antibody trastuzumab (e.g., HERCEPTIN®, Genentech).
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits ErbB2-binding by the anti-ErbB2 antibody trastuzumab.
  • the antibody in the MRD-containing antibody is the anti-ErbB2 antibody trastuzumab.
  • the antibody in the MRD-containing antibody inhibits HER2 dimerization.
  • the antibody in the MRD-containing antibody inhibits HER2 heterodimerization with HER3 (ErbB3).
  • the antibody is pertuzumab (e.g., OMNITARG® and phrMab2C4, Genentech).
  • the antibody specifically binds to the same epitope as pertuzumab.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits binding of ErbB2 by pertuzumab.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • the antibody in the MRD-containing antibody is trastuzumab and 1, 2, 3, 4, 5, 6, or more MRDs in the MRD-containing antibody competitively inhibit binding of ErbB2 by pertuzumab.
  • the antibody in the MRD-containing antibody is an ErbB2-binding antibody selected from the group: MDX-210 (Medarex), tgDCC-E1A (Targeted Genetics), MGAH22 (MacroGenics), and pertuzumab (OMNITARGTM, 2C4; Genentech).
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, or 4 of the above antibodies are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with at least 1, 2, 3 or 4 of the above antibodies.
  • the MRD containing antibody binds ErbB2 and additionally binds ErbB3.
  • the antibody component of the MRD-containing antibody binds ErbB2 and an MRD of the MRD-containing antibody binds ErbB3.
  • the antibody component of the MRD-containing antibody is trastuzumab.
  • the antibody component, MRD component and/or MRD-containing antibody competes for ErbB2-binding with trastuzumab.
  • the antibody in the MRD-containing antibody is an ErbB2-binding antibody selected from MDX-210 (Medarex), tgDCC-E1A (Targeted Genetics), MGAH22 (MacroGenics), and pertuzumab (OMNITARGTM).
  • the antibody component, MRD component and/or MRD-containing antibody competes for ErbB2-binding with an antibody selected from MDX-210, tgDCC-E1A, MGAH22, and pertuzumab.
  • the antibody component of the MRD-containing antibody binds ErbB3 and an MRD of the MRD-containing antibody binds ErbB2.
  • the antibody in the MRD-containing antibody comprises the CDRs of the anti-ErbB2 antibody trastuzumab.
  • the CDR, VH, and VL sequences of trastuzumab are provided in Table 1.
  • VL-CDR1 RAS QDVNTAVAW (SEQ ID NO: 59) VL-CDR2 S AS FLYS (SEQ ID NO: 60) VL-CDR3 Q Q HYTTPPT (SEQ ID NO: 61) VH-CDR1 GRNIKDTYIH(SEQ ID NO: 62) VH-CDR2 RI YPTN GYTRYADSVKG(SEQ ID NO: 63) VH-CDR3 W GGDGFYAMD Y(SEQ ID NO: 64) VL DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQK PGKAPKLLIYSASFLYSGVPSRFSGSRSGTDFTLTISSL QPEDFATYYCQQHYTTPPTFGQGTKVEIKRT (SEQ ID NO: 65) VH EVQLVESGGGLVQPGGSLRLSCAASGFNIKDTYIHWVRQ APGKGLEWVARIYPT
  • the MRD-containing antibody specifically binds ErbB3 (Her3).
  • the antibody is MM121 (Merrimack Pharmaceuticals) or AMG888 (Amgen).
  • the antibody binds to the same epitope as MM121 or AMG888.
  • the antibody competitively inhibits binding of MM121 or AMG888 to ErbB3.
  • the antibody is AV-203 (AVEO).
  • the antibody binds to the same epitope as AV-203.
  • the antibody competitively inhibits binding of AV-203.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1 or both of the above antibodies are also encompassed by the invention.
  • the MRD-containing antibody specifically binds VEGF (VEGFA).
  • the antibody is bevacizumab (e.g., AVASTIN®, Genentech/Roche). In one embodiment, the antibody binds to the same epitope as bevacizumab. In another embodiment, the antibody competitively inhibits binding of bevacizumab to VEGFA.
  • the MRD-containing antibody is AT001 (Affitech). In one embodiment, the antibody binds to the same epitope as AT001. In another embodiment, the antibody competitively inhibits binding of AT001 to VEGFA.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention. Multivalent and multispecific compositions (e.g., MRD-containing antibodies) having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1 or both of the above antibodies are also encompassed by the invention.
  • the antibody in the MRD-containing antibody comprises the CDRs of the anti-VEGF antibody bevacizumab.
  • the CDR, VH, and VL sequences of bevacizumab are provided in Table 2.
  • VL-CDR1 SASQDISNYLN (SEQ ID NO: 72) VL-CDR2 FTSSLHS (SEQ ID NO: 73) VL-CDR3 QQYSTVPWT (SEQ ID NO: 74) VH-CDR1 GYTFTNYGMN (SEQ ID NO: 75) VH-CDR2 WINTYTGEPTYAADFKR (SEQ ID NO: 76) VH-CDR3 YPHYYGSSHWYFDV (SEQ ID NO: 77) VL DIQMTQSPSSLSASVGDRVTITCSASQDISNYLNWYQQKP GKAPKVLIYFTSSLHSGVPSRFSGSGSGTDFTLTISSLQP EDFATYYCQQYSTVPWTFGQGTKVEIKR (SEQ ID NO: 78) VH EVQLVESGGGLVQPGGSLRLSCAASGYTFTNYGMNWVRQA PGKGLEWVGWINTY
  • the antibody in the MRD-containing antibody specifically binds VEGF.
  • the antibody is bevacizumab (e.g., AVASTIN®, Genentech).
  • the antibody binds to the same epitope as bevacizumab.
  • the antibody competitively inhibits binding of bevacizumab to VEGF.
  • the antibody is r84 (Peregrine) or 2C3 (Peregrine).
  • the antibody binds to the same epitope as r84 or 2C3.
  • the antibody competitively inhibits VEGF binding by r84 or 2C3.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, or 3 of the above antibodies are also encompassed by the invention.
  • an MRD-containing antibody binds VEGF and additionally binds an angiogenic target selected from: VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, EFNa1, EFNa2, ANG1, ANG2, IL6, IL8, IL18, HGF, TIE2, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSE, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR2, VEGFR3, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ v ⁇ 3 integrin, TGFb, MMP2, M
  • Multivalent and multispecific compositions that bind VEGF and at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGF.
  • the antibody component of the MRD-containing antibody is r85, 2C3 or AT001.
  • the antibody component of the MRD-containing antibody is bevacizumab.
  • an MRD-containing antibody binds VEGF and additionally binds a target selected from: IL1 beta, phosphatidylserine, TNFSF11 (RANKL), TNFSF12 (TWEAK), IGF1,2, IGF2, IGF1, DKK1, SDF2, CXC3CL1 (fractalkine), sclerostin and tetanus toxoid and HGF.
  • a target selected from: IL1 beta, phosphatidylserine, TNFSF11 (RANKL), TNFSF12 (TWEAK), IGF1,2, IGF2, IGF1, DKK1, SDF2, CXC3CL1 (fractalkine), sclerostin and tetanus toxoid and HGF.
  • an MRD-containing antibody binds VEGF and additionally binds a target selected from: ErbB3, EGFR, cMet, VEGF, RON (MST1R), DLL4, CDCP1 CD318), NRP1, ROBO4, CD13, CTLA4 (CD152), ICOS (CD278), CD20, CD22, CD30, CD33, CD80 and IL6R.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds VEGF.
  • the antibody component of the MRD-containing antibody is r85, 2C3 or AT001.
  • the antibody component of the MRD-containing antibody is bevacizumab.
  • the MRD-containing antibody specifically binds VEGFR1.
  • the antibody competitively inhibits binding of Aflibercept (Regeneron) to VEGFR1.
  • the antibody in the MRD-containing antibody inhibits VEGFR1 dimerization.
  • An MRD that competes for target binding with Aflibercept is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with Aflibercept are also encompassed by the invention.
  • the MRD-containing antibody specifically binds VEGFR2.
  • the antibody is ramucirumab (e.g., IMC1121B and IMC1C11, ImClone).
  • the antibody in the MRD-containing antibody inhibits VEGFR2 dimerization.
  • the antibody binds to the same epitope as ramucirumab.
  • the antibody competitively inhibits binding of ramucirumab to VEGFR2.
  • the antibody competitively inhibits binding of Aflibercept to VEGFR2.
  • An MRD that competes for target binding with ramucirumab is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with ramucirumab or Aflibercept are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds to an FGF receptor (e.g., FGFR1, FGFR2, FGFR3, or FGFR4).
  • the antibody in the MRD-containing antibody is an antibody that specifically binds to FGFR1 (e.g., FGFR1-IIIC).
  • the antibody is IMC-A1 (lineIone), IL one embodiment, the antibody binds to the same epitope as IMC-A1.
  • the antibody competitively inhibits binding of IMC-A1 to FGFR1.
  • the antibody competitively inhibits binding of FP-1039 (Five Prime) to an FGF ligand of FGFR1.
  • the antibody in the MRD-containing antibody is an antibody that specifically binds to FGFR2 (e.g., FGFR2-IIIB and FGFR2-IIIC).
  • the antibody in the MRD-containing antibody is an antibody that specifically binds to FGFR3.
  • the antibody is IMC-A1 (Imclone).
  • the antibody binds to the same epitope as PRO-001 (ProChon Biotech), R3Mab (Genentech), or 1A6 (Genentech).
  • the antibody competitively inhibits binding of PRO-001 (ProChon Biotech), R3Mab (Genentech), or 1A6 (Genentech).
  • MRD that competes for target binding with one of the above antibodies or ligand traps
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1 or more of the above antibodies or ligand traps are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds CD20.
  • the antibody is rituximab (e.g., RITUXAN®/MABTHERA®, Genentech/Roche/Biogen Idec).
  • the antibody binds to the same epitope as rituximab.
  • the antibody competitively inhibits binding of rituximab to CD20.
  • the antibody is GA101 (Biogen Idec/Roche/Glycart).
  • the antibody binds to the same epitope as GA101.
  • the antibody competitively inhibits binding of GA101 to CD20.
  • the antibody is PF-5,230,895 (SBI-087; Pfizer). In one embodiment, the antibody binds to the same epitope as PF-5,230,895. In another embodiment, the antibody competitively inhibits binding of PF-5,230,895 to CD20. In another specific embodiment, the antibody is ocrelizumab (e.g., 2H7; Genentech/Roche/Biogen Idec). In one embodiment, the antibody binds to the same, epitope as ocrelizumab. In another embodiment, the antibody competitively inhibits binding of ocrelizumab to CD20.
  • PF-5,230,895 SBI-087; Pfizer
  • the antibody binds to the same epitope as PF-5,230,895.
  • the antibody competitively inhibits binding of PF-5,230,895 to CD20.
  • the antibody is ocrelizumab (e.g., 2H7; Genentech/Roche/Biogen Idec).
  • the MRD-containing antibody is selected from: obinutuzumab (e.g., GA101; Biogen Idec/Roche/Glycart), ofatumumab (e.g., ARZERRA® and HuMax-CD20 Genmab), veltuzumab (e.g., IMMU-160, Immunomedics), AME-133 (Applied Molecular Evolution), SGN35 (Millennium), TG-20 (GTC Biotherapeutics), afutuzumab (Hoffman-La Roche) and PRO131921 (Genentech).
  • obinutuzumab e.g., GA101; Biogen Idec/Roche/Glycart
  • ofatumumab e.g., ARZERRA® and HuMax-CD20 Genmab
  • veltuzumab e.g., IMMU-160, Immunomedics
  • AME-133 Applied Molecular Evolution
  • SGN35 Stem
  • the antibody binds to the same epitope as an antibody selected from: obinutuzumab, ofatumumab, veltuzumab, AME-133, SGN35, TG-20 and PRO131921.
  • the antibody competitively, inhibits CD20 binding by an antibody selected from obinutuzumab, ofatumumab, veltuzumab, AME-133, SGN35, TG-20, afutuzumab, and PRO131921.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with at least 1, 2, 3, 4, 5, 6 of the above antibodies.
  • an MRD-containing antibody binds CD20 and a target selected from: CD19, CD22, CD30, TNFRSF5 (CD40), CD52, CD74, CD80, CD138, VEGFR1, VEGFR2, EGFR, TNFRSF10A (DR4), TNFRSF10B (DR5), TNF, NGF, VEGF, IGF1,2, IGF2, IGF1 and RANKL.
  • an MRD-containing antibody binds CD20 and a target selected from: CD3, CD4 and NKG2D. Multivalent and multispecific compositions (e.g., MRD-containing antibodies) that bind CD20 and also bind 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD20.
  • the antibody component of the MRD-containing antibody is an antibody selected from: rituximab, GA101, PF-5,230,895, ocrelizumab obinutuzumab, ofatumumab, veltuzumab, AME-133, SGN35, TG-20, afutuzumab and PRO131921.
  • the MRD-containing antibody specifically binds IGF1R.
  • the antibody is selected from cixutumumab (e.g., IMC-A12, Imclone), figitumumab (e.g., CP-751,871, Pfizer), AMG479 (Amgen), BIIB022 (Biogen Idec), SCH 717454 (Schering-Plough), and R1507 (Hoffman La-Roche).
  • the antibody binds to the same epitope as an antibody selected from cixutumumab, figitumumab, AMG479, BIIB022, SCH 717454, and R1507.
  • the antibody competitively inhibits IGF1R binding by an antibody selected from cixutumumab, figitumumab, AMG479, BIIB022, SCH 717454, and R1507.
  • the antibody is figitumumab.
  • the antibody binds to the same epitope as figitumumab.
  • the antibody competitively inhibits IGF1R binding by figitumumab.
  • the antibody is BIIB022.
  • the antibody binds to the same epitope as BIIB022.
  • the antibody competitively inhibits IGF1R binding by BIIB022.
  • the antibody in the MRD-containing antibody inhibits IGF1R dimerization.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for IGF1R binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that compete for IGF1R binding with at least 1, 2, 3, 4, 5, 6 of the above antibodies.
  • an MRD-containing antibody binds IGF1R and a target selected from: EGFR, ErbB2, ErbB3, PDGFRa, PDGFRb, cMet, TNFRSF10A (DR4), TNFRSF10B (DR5), CD20, NKG2D, VEGF, PGE2, IGF1, IGF2 and IGF1,2.
  • an MRD-containing antibody binds IGF1R and a target selected from: CD3, CD4 and NKG2D.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind IGF1R and bind at 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds IGF1R.
  • the antibody component of the MRD-containing antibody is selected from: cixutumumab, figitumumab, AMG479, BIIB022, SCH 717454, and R1507.
  • the multivalent and monovalent multispecific composition binds a target (e.g., ligand, receptor, or accessory protein) associated with an endogenous blood brain barrier (BBB) receptor mediated transport system (e.g., the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and the IGF receptor mediated transport systems) and is capable of crossing to the brain side of the BBB.
  • BBB blood brain barrier
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has 2, 3, 4, 5, or more binding sites (i.e., is capable of multivalently binding) a target antigen (e.g., ligand, receptor, or accessory protein) associated with an endogenous BBB receptor mediated transport system (e.g., the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and the IGF receptor mediated transport systems).
  • a target antigen e.g., ligand, receptor, or accessory protein
  • an endogenous BBB receptor mediated transport system e.g., the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and the IGF receptor mediated transport systems.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has, a single binding site for a target associated with an endogenous BBB receptor mediated transport system.
  • the multivalent and monovalent multispecific composition has 2, 3, 4, 5, or more single binding sites for a target associated with an endogenous BBB receptor mediated transport system.
  • the MRD-containing antibody binds 1, 2, 3, 4, 5, or more targets located on the brain (cerebrospinal fluid) side of the BBB.
  • the MRD-containing antibody additionally binds 1, 2, 3, 4, 5, or more targets located on the brain (cerebrospinal fluid) side of the BBB.
  • the MRD-containing antibody binds 1, 2, 3, 4, 5, or more targets associated with a neurological disease or disorder.
  • the neurological disease or disorder is selected from brain cancer, a neurodegenerative disease, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, ALS, multiple sclerosis, Neuromyelitis optica and Neuro-AIDS (e.g., HIV-associated dementia).
  • the invention encompasses methods of treating a patient by administering a therapeutically effective amount of a multivalent and monovalent multispecific composition to treat a neurological disease or disorder selected from brain cancer, a neurodegenerative disease, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, ALS, multiple sclerosis, Neuromyelitis optica and Neuro-AIDS (e.g., HIV-associated dementia).
  • the multivalent and monovalent multispecific composition is administered to a patient to treat a brain cancer, metastatic cancer of the brain, or primary cancer of the brain.
  • the multivalent and monovalent multispecific composition is administered to a patient to treat a neurological tumor such as, a glioma (e.g., a glioblastoma, glioblastoma multiforme (GBM), and astrocytoma), ependyrnoma, oligodendroglioma, neurofibroma, sarcoma, medulloblastoma, primitive neuroectodermal tumor, pituitary adenoma, neuroblastoma or cancer of the meninges (e.g., meningioma, meningiosarcoma and gliomatosis).
  • the invention encompasses methods of treating a patient by administering a therapeutically effective amount of a multivalent and monovalent multispecific composition to treat a neurodegenerative disorders, or a neurodegenerative
  • the multivalent and monovalent multispecific composition binds an endogenous BBB receptor mediated transport system selected from the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and the IGF receptor mediated transport systems.
  • the multivalent and multispecific composition binds transferrin receptor.
  • the MRD containing antibody binds a target selected from: low-density lipoprotein receptor 1 (LRP-1), a LRP-1 ligand or a functional fragment or variant thereof that binds LRP-1, Low-density lipoprotein receptor 2 (LRP-2), a LRP-2 ligand or a functional fragment or variant thereof that binds LRP-1, a transferrin protein or a functional fragment or variant thereof, insulin receptor, TMEM30A, leptin receptor, IGF receptor, an IGFR ligand or a functional fragment or variant thereof, diphtheria receptor, a diphtheria receptor ligand or a functional fragment or variant thereof, choline transporter, a complex that binds choline receptor, an amino acid transporter (e.g., LAT1/CD98, SLC3A2, and SLC7A5), an amino acid transporter (e.g., LAT1/CD98, SLC3A2,
  • the multivalent and multispecific composition binds RAGE.
  • the multivalent and multispecific composition binds RAGE and a target selected from: Abeta, endothelin1, TNF, IL6, MCSF, an AGE, a S100 member, HMGIB1, LPS and TLR2.
  • Multivalent and multispecific compositions that bind RAGE and also bind 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds RAGE.
  • the multivalent and multispecific composition binds a target antigen associated with an endogenous blood brain barrier (BBB) receptor mediated transport system and also binds a target antigen selected from alpha-synuclein, RGM A, NOGO A, NgR, OMGp MAG, CSPG, neurite inhibiting semaphorins (e.g., Semaphorin 3A and Semaphorin 4) an ephrin, A-beta, AGE (S100 A, amphoterin), NGF, soluble A-B, aggrecan, midkine, neurocan, versican, phosphacan, Te38, and PGE2, ILL IL1R, IL6, IL6R, IL12, IL18, IL23, TNFSF12 (TWEAK), TNFRSF5 (CD40), TNFSF5 (CD40 LIGAND), CD45RB, CD52, CD200, VE
  • the MRD-containing antibody has a single binding site for a target associated with an endogenous blood brain barrier (BBB) receptor mediated transport system and further binds a target selected from alpha-synuclein, RGM A, NOGO A, NgR, OMGp MAG, CSPG, neurite inhibiting semaphorins (e.g., Semaphorin 3A and Semaphorin 4) an ephrin, A-beta, AGE (S100 A, amphoterin), NGF, soluble A-B, aggrecan, midkine, neurocan, versican, phosphacan, Te38, PGE2, IL1, IL1R, IL6, IL6R, IL12, IL18, IL23, TNFSF12 (TWEAK), TNFRSF5 (CD40), TNFSF5 (CD40 LIGAND), CD45RB, CD52, CD200, VEGF, VLA4, TNF alpha, Interfer
  • BBB blood
  • the MRD-containing antibody is administered to a patient to treat a neurological disease or disorder selected from brain cancer, a neurodegenerative disease, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, ALS, multiple sclerosis, Neuromyelitis optica and Neuro-AIDS (e.g., HIV-associated dementia).
  • a neurological disease or disorder selected from brain cancer, a neurodegenerative disease, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, ALS, multiple sclerosis, Neuromyelitis optica and Neuro-AIDS (e.g., HIV-associated dementia).
  • the multivalent and monovalent multispecific composition contains 2 binding sites for 2 or more of the above targets.
  • the multivalent and monovalent multispecific composition contains 2 binding sites for 3 or more targets.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with cancer.
  • the targets bound by the multivalent and monovalent multispecific composition are associated with 1, 2, 3, 4, 5 or more different signaling pathways or modes of action associated with
  • the antibody in the MRD-containing antibody specifically binds integrin.
  • the antibody is selected from: MEDI-522 avb3 (VITAXIN®, MedImmune), CNTO 95 a5b3 (Centocor), JC7U ⁇ v ⁇ 3, and volociximab a5b1 (e.g., M200, PDL and Biogen Idec).
  • the antibody binds to the same epitope as an antibody selected from: MEDI-522, CNTO 95, JC7U ⁇ v ⁇ 3, and volociximab.
  • the antibody competitively inhibits integrin binding by an antibody selected from: MEDI-522, CNTO 95, JC7U, and M200.
  • the antibody is natalizumab (e.g., TSABRI®, Biogen Idec).
  • the antibody binds to the same epitope as natalizumab.
  • the antibody competitively inhibits integrin binding by natalizumab.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds cMet.
  • the antibody is selected from: MetMab (OA-5D5, Genentech), AMG-102 (Amgen) and DN30.
  • the antibody binds to the same epitope as an antibody selected from MetMab), AMG-102 and DN30.
  • the antibody competitively inhibits cMET binding by an antibody selected from MetMab (OA-5D5), AMG-102 and DN30.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, or 3 of the above antibodies are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds cMet and the antibody is selected from: 11E1, CE-355621, LA480 and LMH87. In another embodiment, the antibody binds to the same epitope as an antibody selected from MetMab), AMG-102 and DN30. In another embodiment, the antibody competitively inhibits cMET binding by an antibody selected from: 11E1, CE-355621, LA480 and LMH87.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3 or 4 of the above antibodies are also encompassed by the invention.
  • an MRD containing antibody binds cMET and a target selected from ErbB2, ErbB3, EGFR, IGF1R, NRP1, RON, PDGFRa, PDGFRb, VEGF, VEGFR1, VEGFR2, TGF beta, TGF beta R2, CD82, CD152, NGF, BMP2, BMP4, BMP5, BMP9, BMP10, BMPR-IA, ALK1, a3b1 integrin and HGF.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind cMET and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds cMET.
  • the antibody component of the MRD-containing antibody is an antibody selected from MetMab, AMG-102 and DN30.
  • the antibody component of the MRD-containing antibody is an antibody selected from 11E1, CE-355621, LA480 and LMH87.
  • an MRD-containing antibody binds MST1R (RON).
  • an MRD-containing antibody binds RON and a target selected from EGFR, ErbB2, ErbB3, VEGFR1, VEGFR2, cMET, CXCR4, VEGF, MST, MTSP1, CDCP1, EPHB2, NGF, CXCL12 and HGF (SF).
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds MST1R.
  • the antibody in the MRD-containing antibody specifically binds HGF (SF).
  • the antibody is AMG-102 (Amgen) or SCH 900105 (AV-229, AVEO).
  • the antibody binds to the same epitope as AMG-102 (Amgen) or SCH 900105 (AV-229, AVEO).
  • the antibody competitively inhibits HGF binding by AMG-102 (Amgen) or SCH 900105 (AV-229, AVEO).
  • An MRD that competes for target binding with AMG-102 (Amgen) or SCH 900105 (AV-229, AVEO) is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, or 3 of the above antibodies are also encompassed by the invention.
  • an MRD-containing antibody binds HGF and a target selected from: ErbB2, ErbB3, EGFR, IGF1R, NRP1, RON, PDGFRa, PDGFRb, VEGF, VEGFR1, VEGFR2, TGF beta, TGF beta R2, CD82, CD152, NGF, BMP2, BMP4, BMP5, BMP9, BMP10, BMPR-IA, ALK1, a3b1 integrin, cMET, MST1R (RON), CXCR4, MST, MTSP1, CDCP1, EPHB2, NGF, CXCL12 NRP1 and phosphatidylserine.
  • a target selected from: ErbB2, ErbB3, EGFR, IGF1R, NRP1, RON, PDGFRa, PDGFRb, VEGF, VEGFR1, VEGFR2, TGF beta, TGF beta R2, CD82, CD152, NGF, BMP2, BMP4, BMP5, BMP9, BMP10,
  • Multivalent and multispecific compositions that bind HGF and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds HGF.
  • the antibody component of the MRD-containing antibody is AMG-102 or SCH 900105.
  • the antibody in the MRD-containing antibody specifically binds a5b1 integrin (VLA5).
  • the antibody is volociximab (e.g., M200 Biogen Idec).
  • the antibody binds to the same epitope as volociximab.
  • the antibody competitively inhibits a5b1 integrin binding by volociximab.
  • An MRD that competes for a5b1 integrin binding with volociximab is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for a5b1 integrin binding with volociximab are also encompassed by the invention.
  • the antibody target of the MRD-containing antibody is an antigen associated with an autoimmune disorder, inflammatory or other disorder of the immune system or is associated with regulating an immune response.
  • the MRD-containing antibody improves the performance of antigen presenting cells (e.g., dendritic cells).
  • the antibody target of the MRD-containing antibody is a member selecting from: CD19, CD20, CD21, CD22, CD23, CD27, CD28, CD30, CD30L, TNFSF 14 (LIGHT, HVEM Ligand), CD70, ICOS, ICOSL (B7-H2), CTLA4, PD-1, PDL1 (B7-H1), B7-H4, B7-H3, PDL2 (B7-DC), BTLA, CD46, CD80 (B7-1), CD86 (B7-2), HLA-DR, CD74, PD1, TNFRSF4 (OX40), TNFRSF9 (41BB), TNFSF4 (OX40 Ligand), TNFSF9 (41BB Ligand), TNFRSF1A (TNFR1, p55, p60), TNFRSF1B (TNFR2), TNFRSF13B (T)
  • the antibody target of the MRD-containing antibody is an immunoinhibitory target selected from IL1, IL1 beta, IL1Ra, L-5, IL6, IL6R, CD26L, CD28, CD80, FcRn, and Fc Gamma RIIB.
  • An MRD that binds to one of the above targets is encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • an MRD-containing antibody binds prostaglandin E2 (PGE2).
  • PGE2 prostaglandin E2
  • an MRD-containing antibody binds IL6R and a target selected from: EGFR, IGF1R, IL6R, TNF, NGF, IL1 beta, IL6, IL17A, VEGF, IL15, IL18, S1P and Abeta.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds PGE2.
  • the antibody target of the MRD-containing antibody is an immunostimulatory target (e.g., an agonist of a target associated immune cell activation (such as TNFRSF9 (41BB) or TNFRSF5 (CD40)) or an antagonist of an inhibitory immune checkpoint (such as CTLA-4)).
  • an immunostimulatory target e.g., an agonist of a target associated immune cell activation (such as TNFRSF9 (41BB) or TNFRSF5 (CD40)
  • an antagonist of an inhibitory immune checkpoint such as CTLA-4
  • the antibody target of the MRD-containing antibody is an immunostimulatory target selected from CD25, CD28, CTLA-4, PD1, PDL1, B7-H1, B7-H4, IL10, TGFbeta, TNFSF4 (OX40 Ligand), TNFRSF4 (OX40), TNFSF5 (CD40 Ligand), TNFRSF5 (CD40), TNFSF9 (41BB Ligand), TNFRSF9 (41BB), TNFSF14 (LIGHT, HVEM Ligand), TNFRSF14 (HVEM), TNFSF15 (TL1A), TNFRSF25 (DR3), TNFSF18 (GITR Ligand) and TNFRSF18 (GITR).
  • TNFSF4 OX40 Ligand
  • TNFRSF4 OF40
  • TNFSF5 CD40 Ligand
  • TNFRSF5 CD40
  • TNFSF9 41BB Ligand
  • TNFRSF9 41BB
  • TNFSF14 LIGHT, HVEM Ligand
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the MRD-containing antibody binds 2, 3 or all 4 targets selected from CTLA-4, TNFRSF18 (GITR), 4-1BB, and TNFRSF5 (CD40).
  • the MRD-containing antibody binds CTLA-4 and TNFRSF9 (41BB).
  • the MRD-containing antibody binds CTLA-4 and TNFRSF18 (GITR).
  • the MRD-containing antibody binds CTLA-4 and TNFRSF5 (CD40). In another embodiment, the MRD-containing antibody binds TNFRSF5 (CD40) and TNFRSF9 (41BB). In another embodiment, the MRD-containing antibody binds TNFRSF4 (OX40) and TNFRSF9 (41BB). In another embodiment, the MRD-containing antibody binds PD1 and B7-H1. In an additional embodiment the MRD-containing antibody enhances an immune response, such as the immune system's anti-tumor response or an immune response to a vaccine.
  • the antibody target of the MRD-containing antibody is a cytokine selected from: IL1 alpha, IL1 beta, IL18, TNFSF2 (TNFa), LTalpha, LT beta, TNFSF11 (RANKL), TNFSF13B (13LYS), TNFSF13 (APRIL), IL6, IL7, IL10, IL12, IL15, IL17A, IL23, OncoStatinM, TGFbeta, BMP2-15, PDGF (e.g., PDGF-A, PDGF-B, PDGF-CC, PDGF-C, PDGF-D), an FGF family member (e.g., FGF1, FGF2, FGF4, FGF7, FGF8b and FGF19), VEGF (e.g., VEGFA and VEGFB), MIF, and a type I interferon.
  • cytokine selected from: IL1 alpha, IL1 beta, IL18, TNF
  • MRD that binds to one of the above targets.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that bind to at least 1, 2, 3, 4, 5, 6 of the above targets.
  • the antibody target of the MRD-containing antibody is a cytokine selected from: TNF, CD25, CD28, CTLA-4, PD1, PDL1, B7-H1, B7-H4, IL10, TGFbeta, TNFSF4 (OX40 Ligand), TNFRSF4 (OX40), TNFSF5 (CD40 Ligand), TNFRSF5 (CD40), TNFSF9 (41BB Ligand), TNFRSF9 (41BB), TNFSF14 (LIGHT, HVEM Ligand), TNFRSF14 (HVEM), TNFSF15 (TL1A), TNFRSF25 (DR3), TNFSF18 (GITR Ligand), and TNFRSF18 (GITR).
  • TNFSF4 OX40 Ligand
  • TNFRSF4 OF40
  • TNFSF5 CD40 Ligand
  • TNFRSF5 CD40
  • TNFSF9 41BB Ligand
  • TNFRSF9 41BB
  • TNFSF14 LIGHT, HVEM Ligand
  • MRD that binds to one of the above targets.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that bind to at least 1, 2, 3, 4, 5, 6 of the above targets.
  • the antibody target of the MRD-containing antibody is IL1Ra, IL1Rb, IL2, IL3, IL4, IL7, IL10, IL11, IL15, IL16, IL17, IL17A, IL17F, IL18, IL19, IL25, IL32, IL33, interferon beta, SCF, 13CA1/CXCL13, CXCL1, CXCL2, CXCL6, CXCL13, CXCL16, C3AR, C5AR, CXCR1, CXCR2, CCR1, CCR3, CCR7, CCR8, CCR9, CCR10, ChemR23, CCL3, CCL5, CCL11, CCL13, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL24, CCL25, CCL26, CCL27, MPL, GP130, TLR2, TLR3, TLR4, TLR5, TLR7, TLR8, TLR9, TREM
  • MRD that binds to one of the above targets.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that, bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that bind to at least 1, 2, 3, 4, 5, 6 of the above targets.
  • the above antibody and MRD targets and those otherwise described herein are intended to be illustrative and not limiting.
  • the antibody target of the MRD-containing antibody is TNFSF1A (TNF/TNF-alpha), TNFRSF1A (TNFR1, p55, p60), TNFRSF1B (TNFR2), TNFSF7 (CD27 Ligand, CD70), TNFRSF7 (CD27), TNFSF13B (BLYS), TNFSF13 (APRIL), TNFRSF13B (TACI), TNFRSF13C (BAFFR), TNFRSF17 (BCMA), TNF SF15 (TL1A), TNFRSF25 (DR3), TNFSF12 (TWEAK), TNFRSF12 (TWEAKR), TNFSF4 (OX40 Ligand), TNFRSF4 (OX40), TNFSF5 (CD40 Ligand), TNFRSF5 (CD40), IL1, IL1 beta, IL1R, IL2R, IL4-Ra, IL5, IL5R, IL6, IL6R, IL9,
  • MRD that binds to one of the above targets.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that bind to 1, 2, 3, 4, 5, 6, or more of the above targets are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that bind to at least 1, 2, 3, 4, 5, 6 of the above targets.
  • the antibody target of the MRD-containing antibody competes for target binding with: SGN-70 CD70 (Seattle Genetics), SGN-75 CD70 (Seattle Genetics) Belimumab BLYS (e.g., BENLYSTAR®, Human Genome Sciences/GlaxoSmithKline), Atacicept BLYS/APRIL (Merck/Serono), TWEAK (e.g., Biogen mAb), TL1A antibodies of CoGenesys/Teva (e.g., hum11D8, hum25B9, and hum1B4 (U.S. Appl. Publ. No.
  • OX40 mAb OX40 mAb
  • humAb OX40L Genentech
  • rilonacept IL1 trap e.g., ARCALYST®, Regeneron
  • catumaxomab IL1beta e.g., REMOVAB®, Fresenius Biotech GmbH
  • Xoma052 IL1beta Lilly
  • canakinumab IL1beta e.g., ILARIS® (Novartis) and ACZ885 (Novartis)
  • AMG108 IL1R Amgen
  • daclizumab IL2Ra e.g., ZENAPAX®, Hoffman-La Roche
  • basiliximab IL2Ra e.g., SIMULECT®, Novartis
  • AMGN-317 IL4a Amgen
  • mepolizumab IL5 e.g., BOSATRIA®,
  • SGN30 (Seattle (Genetics) and MDX-060 (Medarex), SGN40 CD40 (Seattle Genetics).
  • ANTOVA® CD40 ligand (Biogen Idec), abatacept CD80 CD86 (e.g., ORENCIA®.
  • GITR e.g., TRX518, (Tolerx), AT010 CXCR3 (Affitech), MLN1202 CCR2 (Millennium Pharmaceuticals), AMG-761 CCR4 (Amgen), HGS004 CCR5 (Human Genome Sciences), PRO 140 (Progenics), MDX-1338 CXCR4 (Medarex), CNTO-888 CCL2 (Centocor), ABN912 CCL2 (Novartis), MDX-1100 CXCL10 (Medarex), TB-403 PLGF (BioInvent), natalizumab integrin Alpha4 subunit (e.g., TYSABRI®, Biogen Idec/Elan), vedolizumab integrin A4B7 (e.g., MLN2, Millennium Pharmaceuticals/Takeda), eculizumab C5 Compliment (e.g., SOLIRIS®, Alex
  • MRD that competes for target binding with one of the above antibodies.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with at least 1, 2, 3, 4, 5 or 6 of the above antibodies.
  • the antibody of the MRD-containing antibody is: SGN-70 CD70 (Seattle Genetics), SGN-75 CD70 (Seattle Genetics), Belimumab BLYS (e.g., BENLYSTA®, Human Genome Sciences/GlaxoSmithKline), BIIB023 TWEAK (Biogen Idec), TL1A antibodies of CoGenesys/Teva (e.g., 11D8, 25B9, and 1B4 (U.S. Appl. Publ. No.
  • OX40 mAb OX40 mAb
  • humAb OX40L Genentech
  • catumaxomab IL1beta e.g., REMOVAB®, Fresenius Biotech GmbH
  • canakinumab IL1beta e.g., ILARIS® (Novartis) and ACZ885 (Novartis)
  • AMG108 IL1R Amgen
  • daclizumab IL2Ra e.g., ZENAPAX®, Hoffman-La Roche
  • basiliximab IL2Ra e.g., SIMULECT®, Novartis
  • AMGN-317 IL4a Amgen
  • mepolizumab IL5 e.g., BOSATRIA®, GlaxoSmithKline
  • reslizumab IL5 e.g., SCH55700, Ception Therapeutics
  • IL21 antibody of Novo Nordisk IL21 antibody Zymogenetics (Zymogenetics), IL22RA, antibody of Zymogenetics, IL31 antibody of Zymogenetics, AMG157 TSLP (Amgen), MEDI-545 interferon alpha (Medimmune), MEDI-546 interferon alpha receptor (Medimmune), AMG811 interferon gamma (Amgen), INNO202 interferon gamma (Innogenetics/Advanced Biotherapy), HuZAF interferon-gamma (PDL), AMG557 B7RP1 (Amgen), AMG191 cKit (Amgen), MOR103 GMCSF (MorphoSys), CAM-3001 GMCSFR (Medimmune), tremelimumab CTLA4 (e.g., CP 675,206, Pfizer), iplimumab CTLA4 MDX-010BMS/Medarex), siplizumab CD
  • the antibody target of the MRD-containing antibody competes for target binding with an antibody selected from: oxelumab (e.g., RG4930; Genmab), AMG139 (Amgen), AMG181 (Amgen), CNTO 148 TNF (Medarex), an anti-TNF antibody described in U.S. Pat. No.
  • ABX-IL8 IL8 (Abgenix), an anti-IL-18 antibody disclosed in US Appl. Pub. No. 2005/0147610 (Abbott), hCBE-11 LTBR (Biogen), HuMax-TAC IL-2Ra CD25) (Genmab, see, e.g., Intl. Appl. Publ. No.
  • WO2004045512 MLN01 Beta2 integrin (Xoma), D3H44 ATF (Genentech), MT203 GMCSF (Micromet and Takeda), IFX1/CaCP29 (InflaRx GmbH), CAT-213 Eotaxin l (Cambridge Antibody Technologies), MDDX-018 IL-8 (e.g., HuMax-InflamTM; Medarex), REGN846 IL-4R (Regeneron, see, e.g., US Appl. Pub. No.
  • MRD that competes for target binding with one of the above antibodies
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, 5, 6, or more of the above antibodies are also encompassed by the invention.
  • the invention encompasses MRD-containing antibodies comprising at least 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with at least 1, 2, 3, 4, 5, or 6 of the above antibodies.
  • one of the above-described antibodies is the antibody of the MRD-containing antibody.
  • the antibody in the MRD-containing antibody specifically binds CTLA4.
  • the antibody is tremelimutnab (e.g., CP-675,206, Pfizer).
  • the antibody binds to the same epitope as tremelimumab.
  • the antibody competitively inhibits binding of tremelimumab to CTLA4.
  • the antibody is ipilimumab (e.g., MDX-010, Bristol-Myers Squibb/Medarex). In one embodiment, the antibody binds to the same epitope as ipilimumab.
  • the antibody competitively inhibits binding of ipilimumab to CTLA4.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CTLA4 binding with tremelimumab or ipilimumab are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds TNFSF12 (TWEAK).
  • TWEAK TNFSF12
  • the antibody is the TWEAK antibody of Biogen that has advanced to Phase I clinical trials.
  • the antibody binds to the same epitope as the Biogen TWEAK antibody.
  • the antibody competitively inhibits binding of the Biogen TWEAK antibody to TWEAK.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for TWEAK binding with the Biogen TWEAK antibody are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds IL2Ra (CD25).
  • the antibody is daclizumab (e.g., ZENAPAX®).
  • the antibody binds to the same epitope as daclizumab.
  • the antibody competitively inhibits binding of daclizumab to IL2Ra (CD25).
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for IL2Ra (CD25) binding with daclizumab are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds CD40 (TNFRSF5).
  • the antibody is CP-870893 CD40 (Pfizer).
  • the antibody binds to the same epitope as CP-870893.
  • the antibody competitively inhibits binding, of CP-870893 to CD40.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for CD40 binding with CP-870893 are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds Alpha4 integrin.
  • the antibody is natalizumab (e.g., TYSABRI®; Biogen Idec/Elan).
  • the antibody binds to the same epitope as natalizumab.
  • the antibody competitively inhibits binding of natalizumab to Alpha4 integrin.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that, compete for Alpha4 integrin binding with natalizumab are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds IL22.
  • the antibody is PF-5,212,367 (ILV-094) (Pfizer).
  • the antibody binds to the same epitope as PF-5,212,367.
  • the antibody competitively inhibits binding of PF-5,212,367 to IL22.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for IL22 binding with PF-5,212,367 are also encompassed by the invention.
  • the antibody in the MRD-containing antibody specifically binds MAdCAM.
  • the antibody is PF-547,659 (Pfizer).
  • the antibody binds to the same epitope as PF-547,659.
  • the antibody competitively inhibits binding of PF-547,659 to MAdCAM.
  • Multivalent and multispecific compositions e.g., MRD; containing antibodies
  • the antibody in the MRD-containing antibody specifically binds TNF.
  • the antibody is adalimumab (e.g., HUMIRA®/TRUDEXA®, Abbott).
  • the antibody binds to the same epitope as adalimumab.
  • the antibody competitively inhibits binding of adalimumab to TNF.
  • the antibody is ATN-103 (Pfizer).
  • the antibody binds to the same epitope as ATN-103.
  • the antibody competitively inhibits binding of ATN-103 to INF.
  • the antibody is infliximab.
  • the antibody binds to the same epitope as infliximab. In another embodiment, the antibody competitively inhibits binding of infliximab to TNF. In another specific embodiment, the antibody is selected from certolizumab (e.g., CIMZIA®, UCB), golimumab (e.g., SIMPONITM, Centocor), and AME-527 (Applied Molecular Evolution). In one embodiment, the antibody binds to the same epitope as certolizumab, golimumab, or AME-527. In another embodiment, the antibody competitively inhibits binding of certolizumab, golimumab, or AME-527, to TNF.
  • certolizumab e.g., CIMZIA®, UCB
  • golimumab e.g., SIMPONITM, Centocor
  • AME-527 Applied Molecular Evolution
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, 3, 4, or 5, of the above antibodies are also encompassed by the invention.
  • the antibody in the MRD-containing antibody comprises the CDRs of the anti-TNF antibody adalimumab.
  • the CDR, VH, and VL sequences of adalimumab are provided in Table 3.
  • VL-CDR1 RASQGIRNYLA (SEQ ID NO: 80) VL-CDR2 AASTLQS (SEQ ID NO: 81) VL-CDR3 RYNRAPYT (SEQ ID NO: 82) VH-CDR1 DYAMH (SEQ ID NO: 83) VH-CDR2 AITWNSGHIDYADSVEG (SEQ ID NO: 84) VH-CDR3 VSYLSTASSLDY (SEQ ID NO: 85) VL DIQMTQSPSSLSASVGDRVTITCRASQGIRNYLAWYQQK PGKAPKLLIYAASTLQSCIVPSRFSGSGSGTDFTLTISS LQPEDVATYYCQRYNRAPYTFOQGTKVEIKR (SEQ ID NO: 86) VH EVQLVESGGGLVQPGRSLRLSCAASGFTEDDYAMHWVRQ APGKGLEWVSAITMTNSGHIDYADSVEGRFTISRDNA
  • an MRD-containing antibody binds TNF (i.e., TNF alpha) and additionally binds a target selected from: Te38, IL12, IL12p40, IL13, IL15, IL17, IL18, IL1beta, IL23, MIF, PGE2, PGE4, VEGF, TNFSF11 (RANKL), TNFSF13B (BLYS), GP130, CD22 and CTLA-4.
  • an MRD-containing antibody binds TNF alpha, IL6, and TNFSF13B (BLYS).
  • an MRD-containing antibody binds TNF alpha and TNFSF12 (TWEAK).
  • the MRD-containing antibody binds TNF and INFSF15 (IL1A).
  • an MRD-containing antibody binds TNF and additionally binds a target selected from NOF, SOST (sclerostin), LPA, IL17A, DKK, alpha Vbeta3, IL23p19, IL2, IL2RA (CD25), IL6, IL6R, IL12p40, IL6, IL10, IL21, IL22 and CD20 binds TNF.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind TNF alpha and at least 1,2,3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TNF alpha.
  • the antibody component of the MRD-containing antibody is adalimumab, infliximab certolizumab golimumab, CNTO 148, AME-527 or ATN-103.
  • the target of the antibody of the MRD-containing antibody is IL6.
  • the antibody of the MRD-containing antibody is siltuximab (CNTO328, Centocor), CNTO-136 (Centocor), CDP-6038 (UCB), or AMGN-220 (Amgen).
  • the antibody of the MRD-containing antibody competes with siltuximab (CNTO328, Centocor), CNTO-136 (Centocor), CDP-6038 (UCB), or AMGN-220 (Amgen) for binding to IL6.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2, or more of the above antibodies are also encompassed by the invention.
  • an MRD-containing antibody binds IL6.
  • an MRD-containing antibody binds IL6 and a target selected from: IL1, IL1 beta, IL1Ra, IL5, CD8, TNFRSF5 (CD40), PDL1, IL6R, IL17A, TNF, VEGF, TNFSF11 (RANKL) and PGE2.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds IL6.
  • the antibody component of the MRD-containing antibody is siltuximab, CNTO136, CDP-6038 or AMGN-220.
  • the target of the antibody of the MRD-containing antibody is IL6R.
  • the antibody of the MRD-containing antibody is REGN-88 (Regeneron) or tocilizumab (ACTEMRATM/ROACTEMRATM, Chugai/Roche).
  • the antibody of the MRD-containing antibody competes with siltuximab, REGN-88 (Regeneron) or tocilizumab (ACTEMRATM/ROACTEMRATM, Chugai/Roche) for binding to IL6R.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1 or both of the above antibodies are also encompassed by the invention.
  • an MRD-containing antibody binds IL6R.
  • an MRD-containing antibody binds IL6R and a target selected from: CD8, TNFRSF5 (CD40), PDL1, IL6, IL17A, TNF, VEGF, TNFSF11 (RANKL) and PGE2.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds IL6R.
  • the antibody component of the MRD-containing antibody is REGN-88 or tocilizumab.
  • an MRD-containing antibody binds TNFSF15 (TL1A).
  • the MRD-containing antibody binds TL1A and a target selected from TNF, IFN alpha, IFN gamma, IL1, IL1beta, IL6, IL8, IL12, IL15, IL17, IL18, IL23 and IL32.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • bind TL1A and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention. These compositions have applications in treating diseases and disorders including inflammatory bowel disease and autoimmune diseases such as rheumatoid arthritis.
  • the antibody component of the MRD-containing antibody binds TL1a.
  • an MRD-containing antibody binds interferon alpha.
  • the MRD-containing antibody binds interferon alpha and TNFSF13B (BLYS).
  • the MRD-containing antibody binds interferon alpha, TNFSF13B (BLYS), and a neutrophil extracellular trap (NET).
  • autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematous.
  • the antibody component of the MRD-containing antibody binds interferon alpha.
  • the multivalent and multispecific compositions of the invention also have applications in treating neurologic diseases or disorders including neurodegenerative diseases, pain and neural injury or trauma.
  • the target of the antibody of the MRD-containing antibody is: amyloid beta (Abeta), beta amyloid, complement factor D, PLP, ROBO4, ROBO, GDNF, NGF, LINGO, or myostatin.
  • the antibody in the MRD-containing antibody is gantenerumab (e.g., R1450, Hoffman La-Roche), bapineuzumab beta amyloid 9 (Elan and Pfizer), solanezumab beta amyloid 9 (Eli Lilly), tanezumab NGF (e.g., RN624, Pfizer), BIIIB033 LINGO (Biogen Idec), PF-3,446,879 myostatin (Pfizer), or stamulumab myostatin (Wyeth).
  • gantenerumab e.g., R1450, Hoffman La-Roche
  • bapineuzumab beta amyloid 9 Eplan and Pfizer
  • solanezumab beta amyloid 9 Eli Lilly
  • tanezumab NGF e.g., RN624, Pfizer
  • BIIIB033 LINGO Biogen Idec
  • PF-3,446,879 myostatin
  • the antibody specifically binds to the same epitope as gantenerumab, bapineuzumab, solarezumab, tanezumab, the Biogen LINGO antibody, or stamulumab.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits target binding by gantenerumab, bapineuzumab, solarezumab, tanezumab, BIIIB033, or stamulumab.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • the target of the antibody of the MRD-containing antibody is beta amyloid.
  • the antibody in the MRD-containing antibody is RN1219 (PF-4,360,365; Pfizer).
  • the antibody specifically binds to the same epitope as RN1219.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits beta amyloid binding by RN 1219.
  • An MRD that competes for beta amyloid binding with RN1219 is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for beta amyloid binding with RN1219 are also encompassed by the invention.
  • the target of the antibody of the MRD-containing antibody is NGF.
  • the antibody in the MRD-containing antibody is tanezumab (e.g., RN624, Pfizer).
  • the antibody specifically binds to the same epitope as tanezumab.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits NGF binding by tanezumab.
  • An MRD that competes for NGF binding with tanezumab is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for NGF binding with tanezumab are also encompassed by the invention.
  • an MRD-containing antibody binds NGF and a target selected from: MTX, NKG2D, RON, IL6R, ErbB3, TNFRSF21 (DR6), CD3, IGFR, DLL4, P1GF, CD20, EGFR, HER2, CD19, CD22, TNFRSF5 (CD40), CD80, cMET, NRP1, TNF, LINGO, HGF, IGF1, IGF1,2, IGF2, NGF, Te38, NogoA, RGM A, MAG, OMGp, NgR, TNFSF12 (TWEAK), PGE2, IL1 beta, Semaphorin 3A and Semaphorin 4.
  • a target selected from: MTX, NKG2D, RON, IL6R, ErbB3, TNFRSF21 (DR6), CD3, IGFR, DLL4, P1GF, CD20, EGFR, HER2, CD19, CD22, TNFRSF5 (CD40), CD80, cMET
  • Multivalent and multispecific compositions that bind NGF and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds NGF.
  • the antibody component of the MRD-containing antibody is tanezumab.
  • the antibody component of the MRD-containing antibody competes for NGF binding with tanezumab.
  • the antibody component of the MRD-containing antibody is MEDI-578.
  • the antibody component of the MRD-containing antibody competes for NGF binding with MEDI-578.
  • the target of the antibody of the MRD-containing antibody is LINGO (e.g., LINGO1).
  • the antibody in the MRD-containing antibody is BIIB033 (Biogen Idec).
  • the antibody specifically binds to the same epitope as BIIB033.
  • the antibody in the MRD containing antibody is an antibody that competitively inhibits LINGO binding by BIIB033.
  • An MRD that competes for LINGO binding with BIIB033 is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for LINGO binding with BIIB033 are also encompassed by the invention.
  • an MRD-containing antibody binds LINGO and a target selected from: MTX, NKG2D, RON, IL6R, ErbB3, TNFRSF21 (DR6), CD3, IGFR, DLL4, P1GF, CD20, EGFR, HER2, CD19, CD22, TNFRSF5 (CD40), CD80, cMET, NRP1, TNF, TNFSF12 (TWEAK), HGF, IGF1, IGF1,2, IGF2, NGF, Te38, NogoA, RGM A, MAG, OMGp, NgR, NGF, PGE2, IL1 beta, Semaphorin 3A and Semaphorin 4.
  • a target selected from: MTX, NKG2D, RON, IL6R, ErbB3, TNFRSF21 (DR6), CD3, IGFR, DLL4, P1GF, CD20, EGFR, HER2, CD19, CD22, TNFRSF5 (CD40), CD80, cMET
  • Multivalent and multispecific compositions that bind LINGO and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds LINGO.
  • the antibody component of, the MRD-containing antibody is BIIB033.
  • the target of an antibody of an MRD-containing antibody is TNFSF12 (TWEAK).
  • TNFSF12 TWEAK
  • the antibody in the MRD-containing antibody binds TNFSF12 (TWEAK) and a target selected from: MTX, NKG2D, RON, IL6R, ErbB3, TNFRSF21 (DR6), CD3, IGFR, DLL4, P1GF, CD20, EGFR, HER2, CD19, CD22, TNFRSF5 (CD40), CD80, cMET, NRP1, TNF, LINGO, HGF, IGF1, GF1,2, IGF2, NGF, Te38, NogoA, RGM A, MAG, OMGp, NgR, NGF, PGE2, IL1 beta, Semaphorin 3A and Semaphorin 4.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind TNFSF12 (TWEAK) and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TNFSF12 (TWEAK).
  • the antibody component of the MRD-containing antibody is BIIB023.
  • the target of the antibody of the MRD-containing antibody is: oxidized LDL, gpIIB, gpIIIa, PCSK9, Factor VIII, integrin a2bB3, AOC3, or mesothelin.
  • the antibody in the MRD-containing antibody is BI-204 oxidized LDL (Bioinvent), abciximab gpIIB, gpIIIa (e.g., REOPRO, Eli Lilly), AMG-145 PCSK9 (Amgen), TB-402 Factor VIII (Bioinvent), vapaliximab, or tadocizumab integrin a2bB3 (Yamonochi Pharma).
  • the antibody specifically binds to the same epitope as BI-204, abciximab, AMG-145, TB-402, or tadocizumab.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits binding of BI-204, abciximab, AMG-145, TB-402, vapaliximab, or tadocizumab.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • the antibody of the MRD-containing antibody is associated with bone growth and/or metabolism.
  • the antibody target of the MRD-containing antibody is TNFSF11 (RANKL).
  • the antibody target of the MRD-containing antibody is DKK1, osteopontin, cathepsin K, TNFRSF19L (RELT), TNFRSF19 (TROY), or sclerostin (CDP-7851 UCB Celltech).
  • antibody target of the MRD-containing antibody is TNFSF11 (RANKL).
  • the antibody in the MRD-containing antibody is denosumab (e.g., AMG-162, Amgen).
  • the antibody specifically binds to the same epitope as denosumab.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits binding of TNFSF11 (RANKL) by denosumab.
  • the antibody is AMG617 or AMG785 (e.g., CDP7851, Amgen).
  • the antibody specifically binds to the same epitope as AMG617 or AMG785.
  • the antibody in the MRD-containing antibody is an antibody that competitively inhibits binding of sclerostin by AMG617 or AMG785.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • an MRD-containing antibody binds TNFSF11 (RANKL).
  • an MRD-containing antibody binds TNFSF11 and a target selected from: sclerostin (SOST), endothelin-1, DKK1, IL1, IL6, IL7, IL8, IL11, IL17A, MCSF, IGF1, IGF2, IGF1,2 IGF1R, TNF, FGF1, FGF2, FGF4, FGF7, FGF8a, FGF8b, FGF18, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TGF beta, TGF beta R2, BMP2, BMP4, BMP5, BMP9, BMP10, BMPR-IA, PDGF, PDGFRa, PDGFRb PTH, PTH related protein (PTHrP
  • Multivalent and multispecific compositions that bind TNFSF11 and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TNFSF 11.
  • the antibody component of the MRD-containing antibody is denosumab, AMG617 or AMG785.
  • the antibody target of the MRD-containing antibody is a bacterial antigen, a viral antigen, a mycoplasm antigen, a prion antigen, or a parasite antigen (e.g., one infecting a mammal).
  • the target of the antibody of the MRD-containing antibody is a viral antigen.
  • the target of the antibody of the MRD-containing antibody is anthrax, hepatitis b, rabies, Nipah virus, west nile virus, a mengititis virus, or CMV.
  • the antibody of the MRD-containing antibody competes with antigen binding with ABTHRAX® (Human Genome Sciences), exbivirumab, foravirumab, libivimmab, rafivirumab, regavirumab, sevirumab (e.g., MSL-109, Protovir), tuvirumab, raxibacumab, Nipah virus M102.4, or MGAWN1® (MacroGenics) for target binding.
  • ABTHRAX® Human Genome Sciences
  • exbivirumab foravirumab, libivimmab, rafivirumab, regavirumab, sevirumab (e.g., MSL-109, Protovir), tuvirumab, raxibacumab, Nipah virus M102.4, or MGAWN1® (MacroGenics)
  • MSL-109, Protovir e.g., MSL-109
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • the target of the antibody of the MRD-containing antibody is RSV.
  • the antibody of the MRD-containing antibody is motavizumab (e.g., NUMAX®, MEDI-577; MedImmune) or palivizumab RSV fusion f protein (e.g., SYNAGIS®, MedImmune).
  • the antibody of the MRD-containing antibody competes with motavizumab or palivizumab RSV fusion f protein, for target binding.
  • the antibody of the MRD-containing antibody is felvizamab. In other embodiments, the antibody of the MRD-containing antibody competes with felvizumab for target binding.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • the target of the antibody of the MRD-containing antibody is a bacterial or fungal antigen.
  • the antibody of the MRD-containing antibody competes for antigen binding with nebacumab, edobacomab (e.g., E5), tefibazumab (Inhibitex), panobacumab (e.g., KBPA101, Kenta), pagibaximab (e.g., BSYX-A110, Biosynexus), urtoxazumab, or efungumab (e.g., MYCOGRAB®, Novartis).
  • nebacumab edobacomab
  • tefibazumab Inhibitex
  • panobacumab e.g., KBPA101, Kenta
  • pagibaximab e.g., BSYX-A110, Biosynexus
  • the antibody of the MRD-containing antibody is nebacumab, eclobacomab, tefibazumab (Inhibitex), panobacumab, pagibaximab, urtoxazumab, or efungumab,
  • An MRD that competes for target binding with one of the above antibodies is also encompassed by the invention.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies having 1, 2, 3, 4, 5, 6, or more MRDs that compete for target binding with 1, 2 or more of the above antibodies are also encompassed by the invention.
  • the antibody in the MRD-containing antibody is the catalytic antibody 38C2.
  • the antibody binds to the same epitope as 38C2.
  • the antibody competitively inhibits 38C2.
  • Human A33 antigen is a transmembrane glycoprotein of the Ig superfamily. The function of the human A33 antigen in normal and malignant colon tissue is not yet known. However, several properties of the A33 antigen suggest that it is a promising target for immunotherapy of colon cancer.
  • These properties include (i) the highly restricted expression pattern of the A33 antigen, (ii) the expression of large amounts of the A33 antigen on colon cancer cells, (iii) the absence of secreted or shed A33 antigen, (iv) the fact that upon binding of antibody A33 to the A33 antigen, antibody A33 is internalized and sequestered in vesicles, and (v) the targeting of antibody A33 to A33 antigen expressing colon cancer in preliminary clinical studies. Fusion of a MRD directed toward A33 to a catalytic or non-catalytic antibody would increase the therapeutic efficacy of A33 targeting antibodies.
  • the antibody in the MRD-containing antibody binds to a human target protein. In some embodiments, the MRD binds to both a human protein and its ortholog in mouse, rat, rabbit, or hamster.
  • the antibodies in the multivalent and multispecific compositions are able to bind their respective targets when the MRDs are attached to the antibody.
  • the antibody binds its target independently.
  • the antibody is a target agonist.
  • the antibody is a target antagonist.
  • the antibody can be used to localize an MRD-containing antibody to an area where the antibody target is located.
  • antibodies used in the present invention may be prepared by any method known in the art.
  • antibody molecules and multivalent and multispecific compositions e.g., MRD-containing antibodies
  • Monoclonal antibodies that can be used as the antibody component of the multivalent and multispecific compositions can be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature 256:495 (1975).
  • hybridoma methods such as those described by Kohler and Milstein, Nature 256:495 (1975).
  • a mouse, hamster, or other appropriate host animal is immunized as described above to elicit the production by lymphocytes of antibodies that will specifically bind to an immunizing antigen. Lymphocytes can also be immunized in vitro.
  • lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene glycol, to form hybridoma cells that can then be selected away from unfused lymphocytes and myeloma cells.
  • a suitable myeloma cell line using, for example, polyethylene glycol, to form hybridoma cells that can then be selected away from unfused lymphocytes and myeloma cells.
  • Hybridomas that produce monoclonal antibodies directed specifically against a chosen antigen as determined by immunoprecipitation, immunoblotting, or by an in vitro binding assay e.g., radioimmunoassay (RIA); enzyme-linked immunosorbent assay (ELISA)
  • an in vitro binding assay e.g., radioimmunoassay (RIA); enzyme-linked immunosorbent assay (ELISA)
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunosorbent assay
  • the monoclonal antibodies can then be purified from the culture medium or ascites fluid as described for polyclonal antibodies above.
  • monoclonal antibodies can also be made using recombinant DNA methods, for example, as described in U.S. Pat. No. 4,816,567.
  • polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cell, such as by RT-PCR using oligonucleotide primers that specifically amplify the genes encoding the heavy and light chains of the antibody, and their sequence is determined using conventional procedures.
  • the isolated polynucleotides encoding the heavy and light chains are then cloned into suitable expression vectors, which when transfected into host cells such as E.
  • monoclonal antibodies are generated by the host cells.
  • recombinant monoclonal antibodies or antibody fragments having the desired immunoreactivity can be isolated from phage display libraries expressing CDRs of the desired species using techniques known in the art (McCafferty et al., Nature 348:552-554 (1990); Clackson et al., Nature 352:624-628 (1991); and Marks et al., J. Mol. Biol. 222:581-597 (1991)).
  • polynucleotide(s) encoding a monoclonal antibody can further be modified in a number of different ways, using recombinant DNA technology to generate alternative antibodies.
  • polynucleotide sequences that encode one or more MRDs and optionally linkers can be operably fused, for example, to the 5′ or 3′ end of sequence encoding monoclonal antibody sequences.
  • the constant domains of the light and heavy chains of for example, a mouse monoclonal antibody can be substituted (1) for those regions of, for example, a human antibody to generate a chimeric antibody or (2) for a non-immunoglobulin polypeptide to generate a fusion antibody.
  • Techniques for site-directed and high-density mutagenesis of the variable region are known in the art and can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
  • the antibody of the MRD-containing antibody is a human antibody.
  • human antibodies can be directly prepared using various techniques known in the art. Immortalized human B lymphocytes immunized in vitro or isolated from an immunized individual that produce an antibody directed against a target antigen can be generated (See, e.g., Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Roemer et al., J. Immunol. 147 (1):86-95 (1991); and U.S. Pat. Nos. 5,750,373 and 6,787,637).
  • the human antibody can be derived from the “minilocus approach” in which an exogenous Ig locus is mimicked through inclusion of individual genes from the Ig locus (see e.g., U.S. Pat. No. 5,545,807).
  • Methods of preparing a human antibody from a phage library, and optionally optimizing binding affinity are known in the art and described, for example, in Vaughan et al., Nat. Biotech. 14:309-314 (1996); Sheets et al., Proc. Nat'l. Acad. Sci. 95:6157-6162 (1998); Hoogenboom et al., Nat. Biotechnology 23:1105-1116 (2005); Hoogenboom et al., J. Mol.
  • Affinity maturation strategies and chain shuffling strategies (Marks et al., Bio/Technology 10:779-783 (1992) (which is herein incorporated by reference in its entirety) are known in the art and can be employed to generate high affinity human antibodies.
  • Antibodies can also be made in mice that are transgenic for human immunoglobulin genes or fragments of these genes and that are capable, upon immunization, of producing a broad repertoire of human antibodies in the absence of endogenous immunoglobulin production. This approach is described in: Lonberg, Nat. Biotechnol 23:1117-1125 (2005), Green et al., Nature Genet. 7:13-21 (1994), and Lonberg et al., Nature 368:856-859 (1994); U.S. Pat. Nos.
  • Multivalent and multispecific compositions can contain a single linker, multiple linkers, or no linker
  • a MRD may be operably attached (linked) to the antibody directly, or operably attached through an optional linker peptide.
  • a MRD may be operably attached to one or more MRD(s) directly, or operably attached to one or more MRD(s) through one or more optional linker peptide(s).
  • Linkers can be of any size or composition so long as they are able to operably attach an MRD and an antibody such that the MRD enables the MRD containing antibody to bind the MRD target.
  • linkers have about 1 to 20 amino acids, about 1 to 15 amino acids, about 1 to 10 amino acids, about 1 to 5 amino acids, about 2 to 20 amino acids, about 2 to 15 amino acids, about 2 to 10 amino acids, or about 2 to 5 amino acids.
  • the linker can also have about 4 to 15 amino acids.
  • the linker peptide contains a short linker peptide with the sequence GGGS (SEQ ID NO:1), a medium linker peptide with the sequence SSGGGGSGGGGGGSS (SEQ ID NO:2), or a long linker peptide with the sequence SSGGGG SGGGGGGSSRSS (SEQ ID NO:19).
  • the MRD is inserted into the fourth loop in the light chain constant region.
  • the MRD can be inserted between the underlined letters in the following amino acid sequence: RTVAAPSVFIFPPSDEQLKSGTASVV CLLNNFYPREAKVQWKVDK LG TNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVY ACEVTHQGLSLPVTKSFNRGEC (SEQ ID NO:102).
  • the linker can also be a non-peptide linker such as an alkyl linker, or a PEG linker.
  • These alkyl linkers may further be substituted by any non-sterically hindering group such as lower alkyl (e.g., C 1 -C 6 ) lower acyl, halogen (e.g., Cl, Br), CN, NH 2 , phenyl, etc.
  • An exemplary non-peptide linker is a PEG linker.
  • the PEG linker has a molecular weight of about 100 to 5000 kDa, or about 100 to 500 kDa.
  • the linker is a “cleavable linker” facilitating release of an MRD or cytotoxic agent in the cell.
  • a “cleavable linker” facilitating release of an MRD or cytotoxic agent in the cell.
  • an acid-labile linker e.g., hydrazone
  • protease-sensitive linker e.g., peptidase-sensitive
  • photolabile linker e.g., dimethyl linker or disulfide-containing linker
  • an MRD or a cytoxic agent and the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • intracellularly cleaved and “intracellular cleavage” refer to a metabolic process or reaction inside a cell on an antibody-drug conjugate (ADC) whereby the covalent attachment, i.e., linked via a linker between the MRD and cytotoxic agent, MRD and antibody, antibody and cytotoxic agent, or between two MRDs is broken, resulting in the free MRD and/or cytotoxic agent dissociated from the antibody inside the cell.
  • ADC antibody-drug conjugate
  • Linker optimization can be evaluated using the techniques described in Examples 1-18 and techniques otherwise known in the art. Linkers preferably should not disrupt the ability of an MRD and/or an antibody to bind target molecules.
  • multi-specie city and greater multi-valency can be achieved through the fusion of MRDs to antibodies.
  • MRDs of the multivalent and multispecific compositions may be operably linked to an antibody through the peptide's N-terminus or C-terminus.
  • the MRD may be operably linked to the antibody at the C-terminal end of the heavy chain of the antibody, the N-terminal end of the heavy chain of the antibody, the C-terminal end of the light chain of the antibody, or the N-terminal end of the light chain of the antibody.
  • Optimization of the MRD composition, MRD-antibody attachment location and linker composition can be performed using the binding assays described in Examples 1-18 and bioassays and other assays known in the art for the appropriate target related biological activity.
  • an MRD-containing antibody is an MRD-containing antibody described in U.S. Application No. 61/489,249, filed May 24, 2011, which is herein incorporated by reference in its entirety.
  • multivalent and multispecific compositions contain an MRD operably linked to either the antibody heavy chain, the antibody light chain, or both the heavy and the light chain.
  • an MRD-containing antibody contains at least one MRD linked to one of the antibody chain terminals.
  • an MRD-containing antibody of the invention contains at least one MRD operably linked to two of the antibody chain terminals.
  • an MRD-containing antibody contains at least one MRD operably linked to three of the antibody chain terminals.
  • an MRD-containing antibody contains at least one MRD operably attached to each of the four antibody chain terminals (i.e., the N and C terminals of the light chain and the N and C terminals of the heavy chain).
  • the MRD-containing antibody has at least one MRD operably attached to the N-terminus of the light chain. In another specific embodiment, the MRD-containing antibody has at least one MRD operably attached to the N-terminus of the heavy chain. In another specific embodiment, the MRD-containing antibody has at least one MRD operably attached to the C-terminus of the light chain. In another specific embodiment, the MRD-containing antibody has at least one MRD operably attached to the C-terminus of the heavy chain.
  • An MRD-containing antibody can be “multispecific” (e.g., bispecific, trispecific tetraspecific, pentaspecific or of greater multispecificity), meaning that it recognizes and binds to two or more different epitopes present on one or more different antigens (e.g., proteins).
  • multispecific e.g., bispecific, trispecific tetraspecific, pentaspecific or of greater multispecificity
  • an MRD-containing antibody is “monospecific” or “multispecific,” (e.g., bispecific, trispecific, and tetraspecific) refers to the number of different epitopes that the MRD-containing antibody binds.
  • Multispecific antibodies may be specific for different epitopes of a target polypeptide (e.g., as described herein) or may be specific for a target polypeptide as well as for a heterologous epitope, such as a heterologous polypeptide target or solid support material.
  • the present invention contemplates the preparation of mono-, bi-, tri-, tetra-, and penta-specific antibodies as well as antibodies of greater multispecificity.
  • the MRD-containing antibody binds two different epitopes.
  • the MRD-containing antibody binds two different epitopes simultaneously.
  • the MRD-containing antibody binds three different epitopes.
  • the MRD-containing antibody binds three different epitopes simultaneously. In another embodiment, the MRD-containing antibody binds four different epitopes. In an additional embodiment the MRD-containing antibody binds four different epitopes simultaneously. In another embodiment, the MRD-containing antibody binds five different epitopes (see, e.g., FIG. 2D ). In an additional embodiment the MRD-containing antibody binds five different epitopes simultaneously.
  • two MRDs of the MRD-containing antibody bind the same antigen. In other embodiments three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same antigen. In other embodiments at least two MRDs of the MRD-containing antibody bind the same antigen. In other embodiments at least three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same antigen. In other embodiments two MRDs of the MRD-containing antibody bind the same epitope. In other embodiments three, four, five, six, seven, eight, nine or ten MRDs of the MRD containing antibody bind the same epitope.
  • At least two MRDs of the MRD-containing antibody bind the same epitope. In other embodiments at least three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same epitope.
  • the antibody and one MRD of the MRD-containing antibody bind the same antigen. In other embodiments the antibody and two, three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same antigen. In other embodiments, the antibody and at least one MRD of the MRD-containing antibody bind the same antigen. In other embodiments the antibody and at least two, three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same antigen. In other embodiments, the antibody and one MRD of the MRD-containing antibody bind the same epitope.
  • the antibody and two, three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same epitope. In other embodiments, the antibody and at least one MRD of the MRD-containing antibody bind the same epitope. In other embodiments the antibody and at least two, three, four, five, six, seven, eight, nine or ten MRDs of the MRD-containing antibody bind the same epitope.
  • the present invention also provides for two or more MRDs which are linked to any terminal end of the antibody.
  • two, three, four, or more MRDs are operably linked to the N-terminal of the heavy chain.
  • two, three, four, or more MRDs are operably linked to the N-terminal of the light chain.
  • two, three, four, or more MRDs are operably linked to the C-terminal of the heavy chain.
  • two, three, four, or more MRDs are operably linked to the C-terminal of the light chain. It is envisioned that these MRDs can be the same or different. In addition, any combination of MRD number and linkages can be used.
  • two MRDs can be operably linked to the N-terminal of the heavy chain of an antibody which contains one MRD linked to the C-terminal of the light chain.
  • three MRDs can be operably linked to the C-terminal of the light chain and two MRDs can be operably linked to the N-terminal of the light chain.
  • Multivalent and multispecific compositions can contain one, two, three, four, five, six, seven, eight, nine, ten or more than ten MRDs.
  • the multivalent and monovalent multispecific composition contains one MRD (see, e.g., FIGS. 2B and 2C ). In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains two MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains three MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains four MRDs (see, e.g., FIGS. 2B and 2C ). In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains five MRDs. In another embodiment, the multivalent and monovalent multispecific composition MRD-containing antibody) contains six MRDs. In an additional embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains between two and ten MRDs.
  • the multivalent and monovalent multispecific composition contains at least one MRD. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least two MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least three MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least four MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least five MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least six MRDs.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains two different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains three different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains four different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains five different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains six different MRDs. In an additional embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains between two and ten different MRDs.
  • the multivalent and monovalent multispecific composition contains at least two different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least three different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least four different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least live different MRDs. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least six different MRDs.
  • the multivalent and multispecific compositions can be MRD monomeric (i.e., containing one MRD at the terminus of a peptide chain optionally connected by a linker) or MRD multimeric (i.e., containing more than one MRD in tandem optionally connected by a linker).
  • the multimeric, multivalent and multispecific compositions can be homo-multimeric (i.e., containing more than one of the same MRD in tandem optionally connected by linker(s) (e.g., homodimers, homotrimers, homotetramer etc.)) or hetero-multimeric (i.e., containing two or more MRDs in which there are at least two different MRDs optionally connected by linker(s) where all or some of the MRDs linked to a particular terminus are different (e.g., heterodimer, heterotrimer, heterotetramer etc.)).
  • linker(s) e.g., homodimers, homotrimers, homotetramer etc.
  • hetero-multimeric i.e., containing two or more MRDs in which there are at least two different MRDs optionally connected by linker(s) where all or some of the MRDs linked to a particular terminus are different (e.g., heterodimer, heterotrimer, heterote
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains two different monomeric MRDs located at different immunoglobulin termini. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains three different monomeric MRDs located at different immunoglobulin termini. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains four different monomeric MRDs located at different immunoglobulin termini. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains five different monomeric MRDs located at different immunoglobulin termini. In another embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains six different monomeric MRDs located at different immunoglobulin termini.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least one dimeric and one monomeric MRD located at different immunoglobulin termini. In another alternative embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least one homodimeric and one monomeric MRD located at different immunoglobulin termini. In another alternative embodiment, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least one heterodimeric and one monomeric MRD located at different immunoglobulin termini.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least one multimeric and one monomeric MRD located at different immunoglobulin termini.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least one homomultimeric and one monomeric MRD located at different immunoglobulin termini.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) contains at least one heteromultimeric and one monomeric MRD located at different immunoglobulin termini.
  • the multivalent and monovalent multispecific composition contains MRDs operably linked to at least two different immunoglobulin termini.
  • the MRDs fused to at least one of the immunoglobulins are a multimer.
  • the MRDs fused to a least one of the immunoglobulins are a homomultimeric (i.e., more than one of the same MRD operably linked in tandem, optionally linked via a linker).
  • the MRDs fused to at least one of the immunoglobulins are a heteromultimeric (i.e., two or more different MRDs operably linked in tandem, optionally linked via a linker).
  • the MRDs fused to at least one of the immunoglobulins are a dimer. In another embodiment, the MRDs fused to a least one of the immunoglobulins are a homodimer. In another embodiment, the MRDs fused to at least one of the immunoglobulins are a heterodimer.
  • the multiple MRDs can target the same target binding site, or two or more different target binding sites. Where the MRDs bind to different target binding sites, the binding sites may be on the same or different target molecules.
  • the antibody and the MRD in a multivalent and monovalent multispecific composition may bind to the same target molecule or to different target molecules.
  • At least one MRD and the antibody in the multivalent and monovalent multispecific composition can bind to their targets simultaneously.
  • each MRD in the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) and the antibody can bind to its target simultaneously. Therefore, in some embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) binds two, three, four, five, six, seven, eight, nine, ten or more targets simultaneously.
  • a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • MRD-containing antibody monovalent multispecific composition
  • Multivalent and Multispecific Compositions having Monovalent Specificity
  • the multivalent and multispecific compositions (e.g., MRD-containing antibodies) of the invention have a single binding site for (i.e., monovalently bind) a target.
  • the antigen binding domains of an antibody component of a multivalent and monovalent multispecific composition of the invention binds to different target epitopes (i.e., the antibody is bispecific).
  • the term “bispecific antibody” is intended to include any antibody, which has two different binding specificities, i.e. the antibody binds two different epitopes, which may be located on the same target antigen or, more commonly, on different target antigens. Methods for making bispecific antibodies are known in the art. (See, for example, Millstein et al., Nature, 305:537-539 (1983); Traunecker et al., EMBO J.
  • compositions of the invention have immunoglobulin chains in which the CH3 domains have been modified by mutating selected amino acids that interact at the interface between two polypeptides so as to preferentially form a bispecific antibody.
  • the bispecific antibodies can be composed of immunoglobulin chains of the same subclass (e.g., IgG1 or IgG3) or different subclasses (e.g., IgG1 and IgG3, or IgG3 and IgG4)
  • a bispecific antibody component of a multispecific and multivalent composition comprises a T366W mutation in the “knobs chain” and T366S, L368A, Y407V mutations in the “hole chain,” and optionally an additional interchain disulfide bridge between the CH3 domains by, e.g., introducing a Y349C mutation into the “knobs chain” and a E356C mutation or a S354C mutation into the “hole chain;” R409D, K370E mutations in the “knobs chain” and D399K, E357K mutations in the “hole chain;” R409D, K370E mutations in the “knobs chain” and D399K, E357K mutations in the “hole chain;” a T366W mutation in the “knobs chain” and T366S, L368A, Y407V mutations in the “hole chain;” R409
  • a bispecific antibody component of a composition of the invention is an IgG4 antibody or a modified IgG4 antibody, or contains an IgG4 heavy chain or a modified IgG4 heavy chain.
  • IgG4 antibodies are dynamic molecules that undergo Fab arm exchange by swapping an IgG4 heavy chain and attached light chain for a heavy-light chain pair from another IgG4 molecule, thus resulting in bispecific antibodies. Accordingly, Fab arm exchange by swapping of MRD-containing-IgG4 antibodies whether caused in vivo or in vitro under physiologic conditions will lead to bispecific antibody compositions.
  • an IgG4 heavy chain of a composition of the invention contains an S228P substitution.
  • an IgG4 heavy chain of a composition of the invention contains a substitution of the Arg at position 409 (e.g., with Lys, Ala, Thr, Met or Leu), the Phe at position 405 (e.g., with Lys, Ala, Thr, Met or Leu) or the Lys at position 370.
  • the CH3 region of an IgG4 heavy chain of a composition of the invention has been replaced with the CHH3 region of IgG1, IgG2 or IgG3.
  • interactions between one or more MRDs located at the C-termini of distinct heavy chains favor and/or stabilize heterodimers between the heavy chains, or otherwise reduces Fab arm exchange by the heterodimer.
  • Exemplary bispecific antibody components of multivalent and multispecific compositions of the invention include, IgG4 and IgG1, IgG4 and IgG2, IgG4 and IgG2, IgG4 and IgG3, IgG1 and IgG3 chain heterodimers.
  • Such heterodimeric heavy chain antibodies can routinely be engineered by, for example, modifying selected amino acids forming the interface of the CH3 domains in human IgG4 and the IgG1 or IgG3 so as to favor heterodimeric heavy chain formation.
  • interactions between one or more MRDs located at the C-termini of heteromeric heavy chains favors or stabilizes heteromultimeric formation or structure, respectively.
  • IgG4 antibodies are known to have decreased ADCC activity and half-life compared to other immunoglobulins subclasses such as, IgG1 and IgG3. Accordingly, IgG4 subclass-based formats provide an attractive format for developing therapeutics that bind to and block cell receptors, but do not deplete the target cell.
  • an IgG4 heavy chain of a composition of the invention can be modified as described herein or otherwise known in the art, so as to increase effector function (e.g., modification of the residues at positions 327, 330 and 331; numbering according to EU index of Kabat).
  • an IgG4 heavy chain of a composition of the invention can be engineered as described herein, or otherwise known in the art to more selectively bind the FcRn at pH 6.0, but not pH 7.4, by for example, incorporating mutations located at the interface between the CH2 and CH3 domains, such as substitutions at T250Q/M428L as well as M252Y/S254T/T256E and H433K/N434F (numbering according to the EU index of Kabat).
  • the multivalent and multispecific compositions (e.g., MRD-containing antibodies) of the invention have a single binding site for (i.e., monovalently bind) a target.
  • the single binding site i.e., monovalent binding site
  • the single binding site is an antibody antigen binding domain.
  • the single binding site is an MRD.
  • the multivalent and multispecific compositions of the invention encompass (and can be routinely engineered to include) MRD-containing antibodies that that contain 1, 2, 3, 4 or more single binding sites for a target.
  • the single binding site(s) may be provided by one or more MRDs located at any one or more of the 4 immunoglobulin heavy chain termini or 4 immunoglobulin light chain termini.
  • single binding site may be provided by one of the antigen binding domains of the antibody (wherein an MRD of the MRD-containing antibody binds the same target epitope of the other antigen binding domain of the antibody.
  • the compositions of the invention encompass (and can be routinely engineered to include) MRD-containing antibodies that contain 1, 2, 3, 4 or more single binding sites for a target and do not bivalenty bind another target
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibodies) has a single binding site for (i.e., monovalently binds) a cell surface target that forms multimers (e.g., homomers or heteromers).
  • the single binding site binds a cell surface target that requires multimerization for signaling.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) has a single binding site that binds a cell surface target and inhibits binding of another molecule (such as a ligand) to the cell surface target.
  • binding of the single binding site inhibits multimerization of the target (e.g., homomeric and heteromeric multimerization).
  • the composition has single binding sites for different targets (i.e., monovalently binds more than one different target). In some embodiments, the multiple single binding sites of the composition bind targets on the same cell. In additional embodiments, the multiple single binding sites of the composition bind targets on different cells. Numerous receptors are known in the art that require multimerization for affecting their normal function. Such receptors are envisioned to be targets of single binding sites in the multivalent and multispecific compositions (e.g., MRD-containing antibodies) of the invention. In some, embodiments, the composition has a single binding site for a receptor tyrosine kinase. In some embodiments, the composition has a single binding site for a growth factor receptor.
  • the composition has a single binding site for a G protein coupled receptor. In additional embodiments the composition has a single binding site for a chemokine receptor. In other embodiments, the composition has a single binding site for a TNF receptor superfamily member.
  • the composition has a single binding site for a receptor selected from: RAGE, c-Met, ErbB2, VEGFR1, VEGFR2, VEGFR3, FGFR1 (e.g., FGFR1-1HC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, PDGFRA, PDGFRB, netrin, CD28, TNFRSF1A (TNFR1, p55, p60), TNFRSF1B (TNFR2), TNFSF6 (Fas Ligand), TNFRSF6 (Fas, CD95), TNFRSF21 or TNFRSF25, TNFRSF7 (CD27), TNFSF8 (CD30 Ligand), TNFRSF8 (CD30), TNFST11 (RANKL), TNFRSF11A (RANK), TNFRSF21 (DR6), TNFRSF25 (DR3), and LRP6.
  • a receptor selected from: RAGE
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) has a single binding site for (i.e., monovalently binds) a cell surface target that forms a multimer and multiple sites (i.e., multivalently binds) for two or more different targets.
  • the multivalent and monovalent multispecific composition has a single binding site for a cell surface target and multiple binding sites for 1, 2, 3, 4, 5 or more different targets.
  • at least 1, 2, 3, 4, 5 or more of the targets bound by the multivalent and monovalent multispecific composition are located on a cell surface.
  • the targets bound by the multivalent and monovalent multispecific composition are soluble targets (e.g., chemokines, cytokines, and growth factors).
  • the composition binds 1, 2, 3, 4, 5 or more of the targets described herein.
  • the targets bound by the composition are tumor antigens (including tumor antigens and tumor associated antigens).
  • a target bound by the composition is associated with a disease or disorder of the immune system.
  • a targets bound by the composition is associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • an MRD-containing antibody has a single binding site for TNFRSF21 (DR6).
  • the MRD-containing antibody has a single binding site for DR6 and binds a target selected from: AGE (S100 A, amphoterin), IL1, IL6, IL18, IL12, IL23, TNFSF12 (TWEAK), TNF alpha, VEGF, TNFRSF5 (CD40), TNFSF5 (CD40 LIGAND), interferon gamma, GMCSF, an FGF, CXCL13, MCP 1, CCR2, NogoA, RGM A, OMgp MAG, a CPSG, LINGO, alpha-synuclein, a semaphorin (e.g., Semaphorin 3A, Semaphorin 4), an ephrin, VLA4, CD45, RB, C5, CD52 and CD200.
  • AGE S100 A, amphoterin
  • Multivalent and multispecific compositions that bind DR6 and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention. These compositions have applications in treating diseases and disorders including neurological diseases and disorders such as multiple sclerosis and other neurodegenerative diseases.
  • the antibody component of the MRD-containing antibody binds DR6.
  • an MRD-containing antibody has a single binding site for TNFRSF25 (DR3).
  • the MRD-containing antibody has a single binding site for DR3 and binds a target selected from: TNF, IFN alpha, IFN gamma, IL1, IL1beta, IL6, IL8, IL12, IL15, IL17, IL18, IL23 and IL32.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • these compositions have applications in treating diseases and disorders including inflammatory bowel disease and autoimmune diseases such as rheumatoid arthritis.
  • the antibody component of the MRD-containing antibody binds DR3.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibodies) has multiple binding site for (i.e., multivalently binds) a cell surface target that forms multimers (e.g., homomers or heteromers).
  • the multiple binding sites bind a cell surface target that requires multimerization for signaling.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) has multiple binding sites for a cell surface target.
  • binding of the multiple binding sites result in multimerization of the target (e.g., homomeric and heteromeric multimerization).
  • the composition has multiple binding sites for different tai gets (i.e., multivalently binds more than one different target).
  • the multiple single binding sites of the composition bind targets on the same cell.
  • the multiple single binding sites of the composition bind targets on different cells.
  • Numerous receptors are known in the art that require multimerization for affecting their normal function. Such receptors are envisioned to be targets of the multivalent and multispecific compositions (e.g., MRD-containing antibodies).
  • the composition has multiple binding sites for a receptor tyrosine kinase.
  • the composition has a multiple binding site for a growth factor receptor.
  • the composition has multiple binding sites for a G protein coupled receptor.
  • the composition has multiple binding sites for a chemokine receptor.
  • the composition has multiple binding, sites for a TNF receptor superfamily member.
  • an MRD-containing antibody binds TNFRSF10A (DR4).
  • the MRD-containing antibody binds DR4 and a target selected from: ErbB2, EGFR, IGF1R, TNFRSF10b (DR5), CD19, CD20, CD22, CD30, CD33, TNFRSF5 (CD40), TNFRSF9 (41BB), IL6, and IGF1,2.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind DR4 and also bind at least 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • compositions have applications in treating diseases and disorders including cancers such as breast cancer, colorectal cancer, head and neck cancer, B-cell lymphomas, hairy cell leukemia, B-cell chronic lymphocytic leukemia and melanoma.
  • cancers such as breast cancer, colorectal cancer, head and neck cancer, B-cell lymphomas, hairy cell leukemia, B-cell chronic lymphocytic leukemia and melanoma.
  • the antibody component of the MRD-containing antibody binds DR4.
  • the antibody component of the MRD-containing antibody is CS1008 or mapatumumab.
  • an MRD-containing antibody binds TNFRSF10B (DR5).
  • an MRD-containing antibody binds DR5 and a target selected from: ErbB2, EGFR, IGF1R, TNFRSF10A (DR4), CD19, CD20, CD22, CD30, CD33, TNFRSF5 (CD40), TNFRSF9 (41BB), IL6, and IGF1,2.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind DR5 and also bind at least 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • compositions have applications in treating diseases and disorders including cancers such as breast cancer, colorectal cancer, head and neck cancer, B-cell lymphomas, hairy cell leukemia, B-cell chronic lymphocytic leukemia, and melanoma.
  • cancers such as breast cancer, colorectal cancer, head and neck cancer, B-cell lymphomas, hairy cell leukemia, B-cell chronic lymphocytic leukemia, and melanoma.
  • the antibody component of the MRD-containing antibody binds DR5.
  • the antibody component of the MRD-containing antibody is LBY135, AMG66, Apomab, PRO95780, lexatumumab, conatumumab or tigatuzumab.
  • the invention also encompasses multivalent and multispecific compositions such as, multivalent and multispecific compositions (e.g., MRD-containing antibodies) that are capable of juxtaposing host effector cells with cells that are desired to be eliminated (e.g., immune cells, cancer cells, diseased cells, infectious agents, and cells infected with infectious agents).
  • multivalent and multispecific functionalities of the compositions of the invention are particularly well suited for redirecting, host immune responses and provide numerous advantages over alternative multispecific composition platforms under development.
  • the multivalent and monovalent multispecific composition binds (1) a target on a cell, tissue, or infectious agent of interest (e.g., an immune cell or a tumor antigen on a tumor cell) and (2) a target on an effector cell so as to direct an immune response to the cell, tissue, or infectious agent of interest.
  • a target on a cell, tissue, or infectious agent of interest e.g., an immune cell or a tumor antigen on a tumor cell
  • the target(s) to which the multivalent, and monovalent multispecific composition binds can be monomeric or multimeric.
  • the mulitimeric target to which a multivalent and monovalent multispecific composition binds can be homomultimeric or heteromultimeric.
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest.
  • one or more targets bound by the multivalent and monovalent multispecific composition is a tumor antigen (e.g., tumor antigens and tumor/cancer associated antigens).
  • the multivalent and multispecific compositions also have applications in treating diseases and disorders including, but not limited to, diseases of the immune system, skeletal system, cardiovascular system, and nervous system, as well as infectious disease.
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is associated with a disease or disorder of the immune system (for example, a disease or disorder of the immune system disclosed herein, such as inflammation or an autoimmune disease (e.g., rheumatoid arthritis)).
  • a disease or disorder of the immune system disclosed herein such as inflammation or an autoimmune disease (e.g., rheumatoid arthritis)
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is associated with a disease or disorder of the skeletal system (e.g., osteoporosis or another disease or disorder of the skeletal system as disclosed herein).
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is associated with a disease or disorder of the cardiovascular system (e.g., a disease or disorder of the cardiovascular system disclosed herein).
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is associated with a disease or disorder of the nervous system (e.g., a disease or disorder of the nervous system disclosed herein).
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is associated with an infectious agent or disease (e.g., an infectious disease or agent disclosed herein).
  • Effector cells that can be bound by a multivalent and monovalent multispecific composition include, but are not limited to, T cells, monocytes/macrophages, and natural killer cells.
  • the target on a cell to which a multivalent and monovalent multispecific composition (e.g., an MRD-containing, antibody) directs an immune response is a tumor antigen.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing, antibody
  • the multivalent and multispecific compositions of the invention are envisioned to be capable of binding virtually any type of tumor and any type of tumor antigen.
  • Exemplary types of tumors that can be targeted include, but are not limited to, one or more cancers selected from the group: colorectal cancer, esophageal, gastric, head and neck cancer, thyroid cancer, multiple myeloma, renal cancer, pancreatic cancer, lung cancer, biliary cancer, glioma, melanoma, liver cancer, prostate cancer, and urinary bladder cancer breast cancer, ovarian cancer, cervical cancer, and endometrial cancer.
  • Exemplary types of tumors that may be targeted include hematological cancers.
  • Hematological cancers that may be targeted include, but are not limited to, one or more cancers selected from the group Hodgkin's lymphoma, medullary non-Hodgkin's lymphoma, acute lymphoblastic leukemia, lymphocytic leukemia, and chronic myelogenous leukemia, acute myelogenous leukemia.
  • Exemplary tumor antigens include ErbB1, ErbB2, ErbB3, VEGFR1, VEGFR2, EGFRvIII, CD16, CD19, CD20, oncostatin M, PSA, PSMA, integrin avb6, ADAM9, CD22, CD23, CD25, CD28, CD36, CD45, CD46, CD56, CD79a/CD79b, CD103, JAM-3, gp100, ALCAM, PIPA, A33, carboxypeptidease M, E-cadherin, CA125, CDK4, CEA, CTLA-4, RAAG10, transferrin receptor, p-15, GD2, MUM-1, MAGE-1, MAGE-3, KSA, MOC31, MIC-1, EphA2, GAGE-1, GAGE-2, MART, KID31, CD44v3, CD44v6, and ROR1. Additional exemplary tumor antigens are described herein and/or known in the art.
  • the target on a cell to which a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) directs an immune response is an immune cell or an inflammatory cell.
  • the invention encompasses a multivalent and monovalent multispecific composition that binds a tumor antigen that is not expressed on tumor cells themselves, but rather on the surrounding reactive and tumor supporting, non-malignant cells comprising the tumor stroma (i.e., tumor associated antigens).
  • the tumor stroma comprises endothelial cells forming new blood vessels and stromal fibroblasts surrounding the tumor vasculature.
  • a multivalent and monovalent multispecific composition binds a tumor associated antigen on an endothelial cell.
  • a multivalent and monovalent multispecific composition binds a tumor antigen and also binds a tumor associated antigen on a fibroblast cell.
  • a multivalent and monovalent multispecific composition binds a tumor antigen and also binds fibroblast activation protein (FAP).
  • FAP fibroblast activation protein
  • Infectious agents to which a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • infectious agents include, but are not limited to, prokaryotic and eukaryotic cells, viruses (including bacteriophage), foreign objects (e.g., toxins), and infectious organisms such as funghi, and parasites (e.g., mammalian parasites), as described herein and infectious agents associated with infectious diseases described herein.
  • infections agents is also intended to encompass other prokaryotic and eukaryotic cells, viruses (including bacteriophage), foreign objects (e.g., toxins), and infectious organisms such as funghi, and parasites otherwise known in the art.
  • the multivalent and monovalent multispecific composition binds (1) a target on a cell, tissue, or infectious agent of interest (e.g., a tumor antigen on a tumor cell) and (2) has a single binding site for a target on an effector cell so as to direct an immune response to the cell, tissue, or infectious agent of interest.
  • the single binding site is an MRD.
  • the single binding site is an antibody antigen binding domain.
  • binding, of the multivalent and monovalent multispecific composition does not elicit a signal when the composition binds a target on an effector cell.
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest. According to some embodiments, at least 1, 2, 3, 4, 5 or more of the targets of the multivalent and monovalent multispecific composition are located on a cell surface. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is a tumor antigen (e.g., tumor antigens and tumor/cancer associated antigens). In additional embodiments, one or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system. In additional embodiments, one or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • a disease or disorder of the skeletal system e.g., osteoporosis
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds (1) a target on a cell, tissue, or infectious agent of interest (e.g., a tumor antigen on a tumor cell) and (2) a target on a leukocyte so as to direct an immune response to the cell, tissue, or infectious agent of interest.
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest. According to some embodiments, at least 1, 2, 3, 4, 5 or more of the targets of the multivalent and monovalent multispecific composition are located on a cell surface.
  • the multivalent and monovalent multispecific composition binds 1, 2, 3, 4, 5 or more targets described herein.
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are a tumor antigen (e.g., tumor antigens and tumor/cancer associated antigens).
  • one or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system.
  • one or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • the invention also encompasses multivalent and multispecific compositions that bind a target expressed on a leukocyte.
  • the multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest.
  • At least 1, 2, 3, 4, 5 or more of the targets of the multivalent and monovalent multispecific composition are located on a cell surface.
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system.
  • 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • the multivalent and monovalent multispecific composition binds a target expressed on a T cell.
  • the multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • the multivalent and monovalent multispecific composition has multiple binding sites for (i.e., multivalently binds) a target on a T cell.
  • the multivalent and monovalent multispecific composition has a single binding site for (i.e., monovalently binds) a target on a T cell.
  • the single binding site is an MRD.
  • the single binding site is an antibody antigen binding domain.
  • binding of the multivalent and monovalent multispecific composition does not elicit a signal when the composition binds a target on a T cell.
  • the binding of the multivalent and monovalent multispecific composition does not result in lysis of the T cell expressing the target.
  • the multivalent and monovalent multispecific composition binds a target selected from: CD2, CD3, CD4, CD8, CD161, a chemokine receptor. CD5, and CCR5.
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest. According to some embodiments, at least 1, 2, 3, 4, 5 or more of the targets of the multivalent and monovalent multispecific composition are located on a cell surface. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition is a tumor antigen (e.g., tumor antigens and tumor/cancer associated antigens). In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • a disease or disorder of the skeletal system e.g., osteoporosis
  • the multivalent and monovalent multispecific composition contains a fusion protein containing one or more peptides that bind to a protein on the surface of a cell, such as a T cell.
  • the multivalent and monovalent multispecific composition bind target membrane proximal protein sequences on a cell and inhibit the cross-linking (e.g., multimerization) of the target protein or its associated proteins.
  • the multivalent and monovalent multispecific composition binds to a T cell and inhibits the cross-linking of the cell protein or its associated proteins.
  • the multivalent and multispecific antibody comprises the amino terminal 27 amino acids of mature CD3 epsilon.
  • the multivalent and monovalent multispecific composition comprises a fusion protein containing one or more proteins corresponding to the G Domain of a CD3 protein (e.g., CD3 epsilon, CD3 gamma, CD3 alpha (TCRA) or CD3 beta (TCRB).
  • a CD3 protein e.g., CD3 epsilon, CD3 gamma, CD3 alpha (TCRA) or CD3 beta (TCRB).
  • the fusion protein comprises a polypeptide having an amino acid sequence selected from GYYVCYPRGSKPEDANFYLYLR ARVC (SEQ ID NO:21), YLYLRAR (SEQ ID NO:22), YRCNGTDIYKDKESTVQ VHYRMC (SEQ ID NO:23), and DKESTVQVH (SEQ ID NO:24).
  • the composition comprises a fusion protein containing one or more proteins corresponding to a portion of the extracellular domain of a CD3 protein (e.g., CD3 epsilon, CD3 gamma, CD3 alpha (TCRA) or CD3 beta (TCRB)) that is able to bind CD3, or a CD3 multimer.
  • a CD3 protein e.g., CD3 epsilon, CD3 gamma, CD3 alpha (TCRA) or CD3 beta (TCRB)
  • TCRA CD3 alpha
  • TCRB CD3 beta
  • the fusion protein comprises a portion of a CD3 protein that is able to bind CD3 or a CD3 multimer wherein the portion comprises a CD3 binding fragment of a polypeptide having an amino acid sequence selected from: KIPIEELEDRVFVNCNTSITWVEG TVGTLLSDITRLDLGKRILDPRGIYRCNGTDIY KDKESTVQVHYRMCQSCVELD (human CD3 delta mature ECD, SEQ ID NO:25), QSIKGNHLVKVYDYQEDGSVLLTCDAEAK NITWFKDGKMIGFLTEDKKKWNLGSNAKDPRGMYQCKGSQNKSKPLQVYRMCQNC IELN (human CD3 gamma matte ECD, Ig-like domain highlighted; SEQ ID NO:26), GNEEMGGITQTPYKVSTSGTTTVILTCPQYPGSEILWQHNDKNIGGDEDDKNIGS DEDHL SLKEFSELEQSGYYVCYPRGS
  • the chemokine fragment is a portion of a chemokine selected from: CCL20 (LARC/Ck ⁇ 4), CCL25 (TECK/Ck ⁇ 15), CXCL12 (SDF-1), CXCL13 (BCA-1), CXCL16 (SRPSOX), and CX3CL1 (Fractalkine).
  • the chemokine fragment is a portion of a chemokine selected from: CCL5 (RANTES), CCL8 (MCP-2), CXCL9 (MIG/CRG-10), CXCL10 (IP-10/CRG-2) and CXCL11 (TAC/IP-9).
  • the chemokine fragment is a portion of a chemokine selected from CCL3 (MIP-1a) and CCL4 (MIP-1 ⁇ ).
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds CD3.
  • the composition binds a CD3 target selected from CD3 delta, CD3 epsilon, CD3 gamma, CD3 zeta, TCR alpha, TCR beta, the TCR complex, or a heteromeric or homomultimeric combination thereof.
  • the composition binds CD3 epsilon.
  • the multivalent and monovalent multispecific composition binds CD3 and multiple binding sites for 1, 2, 3, 4, 5 or more different targets (e.g., a tumor antigen as disclosed herein or otherwise known in the art).
  • the multivalent and monovalent multispecific composition has a single binding site for (i.e., monovalently binds) CD3.
  • the multivalent and monovalent multispecific composition has a single MRD that binds CD3 and multiple binding sites for 1, 2, 3, 4, 5 or more different targets (e.g., a tumor antigen as disclosed herein or otherwise known in the art).
  • the multivalent and monovalent multispecific composition has a single antibody antigen binding domain that binds CD3 and multiple binding sites for 1, 2, 3, 4, 5 or more different targets (e.g., a tumor antigen as disclosed herein or otherwise known in the art).
  • the CD3 binding compositions of the invention are not single chain antibodies.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds human CD3 and a CD3 ortholog from another organism. In additional embodiments, the multivalent and monovalent multispecific composition binds human CD3 and a CD3 ortholog from another primate. In further embodiments, the multivalent and monovalent multispecific composition binds human CD3 and a CD3 ortholog from cynomolgus Monkey or rhesus Monkey. In other embodiments, the multivalent and monovalent multispecific composition binds human CD3 and a CD3 ortholog from a primate selected from Saguinus Oedipus and Callithrix jacchus ).
  • the multivalent and monovalent multispecific composition binds human CD3 and a CD3 ortholog from cynomolgus monkey, and a CD3 ortholog from mouse or rat.
  • the human CD3 epsilon binding compositions of the invention are not single chain antibodies.
  • the CD3 binding compositions of the invention are not single chain antibodies.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds human CD3 epsilon.
  • the, multivalent and monovalent multispecific composition binds human CD3 epsilon protein having the sequence of amino acids 23-207 set forth in NCBI Ref. Seq. No. NP — 000724.
  • the multivalent and monovalent multispecific composition binds a polypeptide having the amino acid sequence of QDGNEEMGGITQTPYKVSISGTT VILT (SEQ ID NO:29).
  • the multivalent and monovalent multispecific composition binds a polypeptide having the amino acid sequence of QDGNEEMGGI (SEQ ID NO:30).
  • the multivalent and monovalent multispecific composition binds a polypeptide having the amino acid sequence of QDGNEEMGG (SEQ ID NO:31).
  • the human CD3 epsilon binding compositions of the invention are not single chain antibodies.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • a single binding site for CD3 epsilon i.e., monovalently binds CD3 epsilon
  • multiple binding sites for 1, 2, 3, 4, 5 or more different targets e.g., a B cell or other target disclosed herein.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) competes for binding to CD3 with an antibody selected from: OKT-3, otelixizumab, teplizumab, visilizumab, muromonab, X35-3, VIT3, BMA030 (BW264/56), CLB-T3/3, CRIS7, YTH12.5, F111409, CLB-T3.4.2, TR-66, WT31, WT32, SPv-T3b, 11D8, XIII-141, XIII46, 12F6, T3/RW2-8C8, T3/RW24B6, OKT3D, M-T301, SMC2 and F101.01.
  • an antibody selected from: OKT-3, otelixizumab, teplizumab, visilizumab, muromonab, X35-3, VIT3, BMA030 (BW264/56), CLB-T3/3, CRIS7, Y
  • an MRD of an MRD-containing antibody competes for binding to CD3 with an antibody selected from OKT-3, otelixizumab, teplizumab, visilizumab, muromonab X35-3, VIT3, BMA030 (BW264/56), CLB-T3/3, CRIS7, YTH12.5, F111409.
  • OKT3D M-T301, SMC2 and F101.01.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) competes for binding to CD3 with a CD3 binding composition disclosed in Int. Appt. Pub Nos. WO2004/106380 and WO99/54440; funnacliffe et al., Int. Immunol. 1:546-550 (1989); Kjer-Nielsen, PNAS 101:7675-7680 (2004); or Salmeron et al., J. Immunol. 147: 3047-3052 (1991).
  • MRD-containing antibody e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition binds human CD3 epsilon and a CD3 epsilon ortholog from another organism. In some embodiments, the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds human CD3 epsilon and a CD3 epsilon ortholog from another primate. In additional embodiments, the multivalent and monovalent multispecific composition binds human CD3 epsilon and a CD3 epsilon ortholog from cynomolgus monkey or rhesus monkey.
  • the multivalent and monovalent multispecific composition binds human CD3 epsilon and a CD3 epsilon ortholog from a primate selected from Saguinus Oedipus and Callithrix jacchus .
  • the multivalent and monovalent multispecific composition binds human CD3 epsilon and a CD3 epsilon ortholog from cynomolgus monkey, and a CD3 epsilon ortholog from mouse or rat.
  • an MRD of the multivalent and monovalent multispecific composition binds CD3 epsilon.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds human CD3 delta.
  • the, multivalent and monovalent multispecific composition binds human CD3 delta having the sequence of amino acids 22-171 set forth in NCBI Ref Seq. No. NP — 000723.
  • an MRD of the multivalent and monovalent multispecific composition binds CD3 delta.
  • an antibody antigen binding domain of the multivalent and monovalent multispecific composition binds CD3 delta.
  • the human CD3 epsilon binding compositions of the invention are not single chain antibodies.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing, antibody) binds human CD3 gamma protein having the sequence of amino acids 23-182 set forth in NCBI Ref. Seq. No. NP — 000064.
  • an MRD of the multivalent and monovalent multispecific composition binds gamma.
  • an MRD of the multivalent and monovalent multispecific composition binds CD3 gamma.
  • an antibody antigen binding domain of the multivalent and monovalent multispecific composition binds CD3 gamma.
  • the human CD3 gamma binding compositions of the invention are not single chain antibodies.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds human CD3 zeta protein having the sequence of amino acids 22-464 set forth in NCBI Ref. Seq. No. NP — 932170.
  • an MRD of the multivalent and monovalent multispecific composition binds CD3 zeta.
  • an antibody antigen binding domain of the multivalent and monovalent multispecific composition binds CD3 zeta.
  • the human CD3 zeta binding compositions of the invention are not single chain antibodies.
  • the invention also encompasses multivalent and multispecific compositions that bind a target expressed on a natural killer cell
  • the multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • the multivalent and monovalent multispecific composition has multiple binding sites for (i.e., monovalently binds) a target on a natural killer cell.
  • the multivalent and monovalent multispecific composition has a single binding site for (i.e., monovalently hinds) a target on a natural killer cell.
  • the single binding site is an MRD. In other embodiments, the single binding site is an antibody antigen binding domain. In further embodiments, binding of the multivalent and monovalent multispecific composition does not elicit a signal when the composition binds a target on a natural killer cell. In some embodiments, the multivalent and monovalent multispecific composition binds a target selected from: KLRD1, KLRK1, KLRB1, 2B4 (CD244), KIR2D4, KIR2D5, and KIR3DL1. In other embodiments, the multivalent and monovalent multispecific composition binds a target selected from: CD56, CD2, and CD161.
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest. According to some embodiments, at least 1, 2, 3, 4, 5 or more of the targets of the multivalent and monovalent multispecific composition are located on a cell surface. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are a tumor antigen (e.g., tumor antigens and tumor/cancer associated antigens). In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • a disease or disorder of the skeletal system e.g., osteoporosis
  • the multivalent and monovalent multispecific composition binds CD2.
  • the multivalent and monovalent multispecific composition binds human CD2.
  • the multivalent and monovalent multispecific composition binds human CD2 protein having the sequence of amino acids 25-209 set forth in NCBI Ref Seq. No. NP — 001758.
  • the multivalent and monovalent multispecific composition has multiple binding sites for CD2.
  • the single binding site is an MRD.
  • the single binding site is an antibody antigen binding domain.
  • the multivalent and monovalent multispecific composition has a single binding site for CD2.
  • binding of the multivalent and monovalent multispecific composition to CD2 does not elicit a signal by the cell on which CD2 is expressed.
  • the multivalent and monovalent multispecific composition binds CD2 and 1, 2, 3, 4, 5 or more different targets (e.g., a tumor antigen as disclosed herein or otherwise known in the art).
  • the CD2 binding compositions of the invention are not single chain antibodies.
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds human CD2 at d a CD2 ortholog from another organism.
  • the multivalent and monovalent multispecific composition binds human CD2 and a (D2 ortholog from another primate.
  • the multivalent and monovalent multispecific composition binds human CD2 and a CD2 ortholog from cynomolgus monkey or rhesus monkey.
  • the multivalent and monovalent multispecific composition binds a target on a myeloid cell.
  • the multivalent and monovalent multispecific composition binds (1) a target on a cell, tissue, or infectious agent of interest (e.g., a tumor antigen on a tumor cell) and (2) a target on an immune accessory cell (e.g., myeloid cell) so as to juxtapose myeloid cells with the cell, tissue, or infectious agent of interest.
  • the multivalent and monovalent multispecific composition has multiple binding sites for (i.e., multivalently binds) a target on a myeloid cell.
  • the multivalent and monovalent multispecific composition has a single binding site for (i.e., monovalently binds) a target on an accessory cell (e.g., myeloid cell).
  • the single binding site is an MRD.
  • the single binding site is an antibody antigen binding domain.
  • binding of the multivalent and monovalent multispecific composition does not elicit a signal when the composition binds a target on a myeloid cell.
  • the multivalent and monovalent multispecific composition binds an Fc gamma receptor selected from CD16 (i.e., Fc gamma RIII), CD64 (i.e., Fc gamma RI), and CD32 (i.e., Fc gamma RII).
  • the multivalent and monovalent multispecific composition binds CD64 (i.e., Fc gamma RI).
  • the multivalent and monovalent multispecific composition binds a target selected from, MHC class 2 and its invariant chain, TLR1, TLR2, TLR4, TLR5 and TLR6.
  • the multivalent and monovalent multispecific composition binds at least 2, 3, 4, or 5 targets on the cell, tissue, or infectious agent of interest. According to some embodiments, at least 1, 2, 3, 4, 5 or more of the targets of the multivalent and monovalent multispecific composition are located on a cell surface. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are a tumor antigen (e.g., tumor antigens and tumor/cancer associated antigens). In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the immune system. In additional embodiments, 1, 2, 3, 4, 5 or more targets bound by the multivalent and monovalent multispecific composition are associated with a disease or disorder of the skeletal system (e.g., osteoporosis), cardiovascular system, nervous system, or an infectious disease.
  • a disease or disorder of the skeletal system e.g., osteoporosis
  • the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds a target of interest on a cancer cell. In additional embodiments, the multivalent and monovalent multispecific composition binds a target of interest on an immune cell. In further embodiments, the multivalent and monovalent multispecific composition binds a target of interest on a diseased cell. In other embodiments, the multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) binds a target of interest on an infectious agent (e.g., a bacterial cell or a virus).
  • an infectious agent e.g., a bacterial cell or a virus
  • the invention encompasses a method of treating a disease or disorder by administering to a patient in need thereof, a therapeutically effective amount of a multivalent and monovalent multispecific composition of the invention.
  • Particular embodiments are directed to a method of treating a disease or disorder by administering to a patient in need thereof, a therapeutically effective amount a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that has a single binding site for a target (i.e., that monovalently binds a target).
  • the administered multivalent and monovalent multispecific composition has a single binding site for a target on a leukocyte, such as a T-cell (e.g., CD3).
  • the administered multivalent and monovalent multispecific composition has a single binding site for a target on a leukocyte, such as a T-cell (e.g., CD3) and multiple binding sites for (i.e., is capable of multivalently binding) a target located on a cell or tissue of interest (e.g., a tumor antigen on a tumor cell).
  • a leukocyte such as a T-cell (e.g., CD3)
  • multiple binding sites for (i.e., is capable of multivalently binding) a target located on a cell or tissue of interest e.g., a tumor antigen on a tumor cell.
  • the invention is directed to treating a disease or disorder by administering to a patient a therapeutically effective amount of a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that has a single binding site for a target (i.e., that monovalently binds a target) and multiple binding sites for 1, 2, 3, 4, 5 or more different targets.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • the invention is directed to treating a disease or disorder by administering to a patient in need thereof, a therapeutically effective amount of a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that has a single binding site for CD3 (e.g., CD3 epsilon) that monovalently binds CD3 and multiple binding sites for 1, 2, 3, 4, 5 or more different targets.
  • a multivalent and monovalent multispecific composition e.g., an MRD-containing antibody
  • CD3 e.g., CD3 epsilon
  • the tumor cell is from a cancer selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • a cancer selected from breast cancer, colorectal cancer, endometrial cancer, kidney (renal cell) cancer, lung cancer, melanoma, Non-Hodgkin Lymphoma, leukemia, prostate cancer, bladder cancer, pancreatic cancer, and thyroid cancer.
  • the MRD(s) and the antibody in the MRD-containing antibody are antagonists of their respective targets. In other embodiments, the MRD(s) and the antibody in the MRD-containing antibody are agonists of their respective target. In yet other embodiments, at least one of the MRDs in the MRD-containing antibody is an antagonist of its target molecule and the antibody is an agonist of its target molecule. In yet another embodiment, at least one of the MRDs in the MRD-containing antibody is an agonist of its target molecule, and the antibody is an antagonist of its target molecule.
  • both the MRD(s) and the antibody in the MRD-containing antibody bind to soluble factors. In some embodiments, both the MRD(s) and the antibody in the MRD-containing antibody bind to cell surface molecules. In some embodiments, at least one MRD in the MRD-containing antibody binds to a cell surface molecule and the antibody in the MRD-containing antibody hinds to a soluble factor. In some embodiments, at least one MRD in the MRD-containing antibody binds to a soluble factor and the antibody in the MRD-containing antibody binds to a cell surface molecule.
  • An improved multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) that specifically binds a desired target or targets can also be prepared based on a previously known MRD or multivalent and monovalent multispecific composition (e.g., MRD-containing antibody).
  • 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 10-20, 20-30, 30-50, 50-100, 100-150 or more than 150 amino acid substitutions, deletions or insertions can be introduced into an MRD or multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) sequence and the resulting MRD or multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) can be screened for binding to the desired target or targets, for antagonizing target activity, or for agonizing target activity as described in the examples or using techniques known in the art.
  • Additional peptide sequences may be added, for example, to enhance the in vivo stability of the MRD or affinity of the MRD for its target.
  • the binding of a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • its target e.g., a cell
  • the binding is at least about 2-fold, at least about 5-fold, at least about 10-fold, at least about 20-fold, at least about 50-fold, at least about 75-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold improved.
  • the binding of a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • a target e.g., a cell or a molecule containing multiple epitopes
  • the binding of the multivalent and monovalent multispecific composition is enhanced compared to the binding of the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) to a target (e.g., a cell or a molecule containing multiple epitopes) expressing only the MRD target or only the antibody target.
  • the binding is at least about 2-fold, at least about 5-fold, at least about 10-fold, at least about 20-fold, at least about 50-fold, at least about 75-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold improved.
  • This increased avidity can enable multivalent and multispecific compositions (e.g., MRD-containing antibodies) to bind to targets that have previously been difficult to target, e.g., G-protein coupled receptors and carbohydrate molecules.
  • the binding of a multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) to an MRD target is enhanced in a region (e.g., of the body) where the antibody target is localized compared to a region where the antibody target is not expressed or is expressed at a lower level.
  • the binding of a multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) to an antibody target is enhanced in a region (e.g., of the body) where the MRD target is localized compared to a region where the MRD target is not expressed or is expressed at a lower level.
  • the binding is at least about 2-fold, at least about 5-fold, at least about 10-fold, at least about 20-fold, at least about 50-fold, at least about 75-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold improved.
  • the multivalent and monovalent multispecific composition retains particular activities of the parent antibody.
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition is capable of inducing complement dependent cytotoxicity.
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • ADCC antibody dependent cell mediated cytotoxicity
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition is capable of reducing tumor volume.
  • the multivalent and multispecific compositions are capable of inhibiting tumor growth.
  • the multivalent and monovalent multispecific composition shows improved activity or pharmacodynamic properties compared to the corresponding antibody without the attached MRD.
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • the MRD-containing antibody has a greater therapeutic efficacy than the corresponding antibody without the attached MRD.
  • the multivalent and multispecific compositions have one or more of the following effects: inhibit proliferation of tumor cells, reduce the tumorigenicity of a tumor, inhibit tumor growth, increase patient survival, trigger cell death of tumor cells, differentiate tumorigenic cells to a non-tumorigenic state, or prevent metastasis of tumor cells.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) is at least as stable as the corresponding antibody without the attached MRD. In certain embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) is more stable than the corresponding antibody without the attached MRD. MRD-antibody stability can be measured using methods known to those in the art, including, for example, ELISA techniques. In some embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) is stable in whole blood at 37° C.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has at least the same affinity for Fc receptors as the corresponding parent antibody. In other nonexclusive embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has at least the same affinity for complement receptors as the corresponding parent antibody. In other nonexclusive embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has at least the same half-life as the corresponding parent antibody. In other embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) can be expressed at levels commensurate with the corresponding parent antibody.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has an increased affinity for Fc receptors compared to the corresponding parent antibody.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has an increased affinity for complement receptors compared to the corresponding parent antibody.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) has an increased half-life compared to the corresponding parent antibody.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) can be expressed at increased levels compared to that of the corresponding, parent antibody.
  • antibody-drug conjugates for the local delivery of cytotoxic agents, allows targeted delivery of the drug to tumors, and intracellular accumulation therein, where systemic administration of these unconjugated drug agents may result in unacceptable levels of toxicity to normal cells as well as the tumor cells sought to be eliminated (Baldwin et al., Lancet pages 603-05 (1986); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review,” in Monoclonal Antibodies '84: Biological And Clinical Applications, A. Pinchera et al., (ed.s), pp. 475-506) (1985)).
  • the invention encompasses a multivalent and monovalent multispecific composition (e.g., an MRD-containing antibody) that is covalently or otherwise associated with a cytotoxic agent (payload) (i.e., as multivalent and monovalent multispecific-cytoxic agent complexes (e.g., MRD-containing antibody-cytoxic agent complexes).
  • a cytotoxic agent payload
  • the cytoxic agent is covalently attached to a multivalent and monovalent multispecific composition (e.g., MRD containing antibody) by a linker.
  • the linker attaching the multivalent and monovalent multispecific composition and the cytotoxic agent is cleavable by a protease.
  • the cytotoxic agent is a chemotherapeutic agent, growth inhibitory agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), a radioactive isotope (i.e., a radioconjugate) or a prodrug.
  • toxin e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof
  • a radioactive isotope i.e., a radioconjugate
  • Methods of using immunoconjugates are also encompassed by the invention.
  • Cytotoxic agents that may be covalently or otherwise associated with multivalent and multispecific compositions include, but are not limited to any agent that is detrimental to (e.g., kills) cells.
  • Cytotoxins useful in the compositions and methods of the invention include, inter alia, alkylating agents intercalating agents, antiproliferative agents, anti-mitototic agents, tubulin binding agents, vinca alkaloids, enediynes, trichothecenes, podophyllotoxins or podophyllotoxin derivatives, the pteridine family of drugs, taxanes, anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin, dolastatins (e.g., dolastatin 10, dolastatin 11, and dolastatin 15)), topoiosomerase inhibitors, and platinum complex chemotherapeutic
  • compositions of the invention include a cytoxic agent that is a tubulin depolymerizing agent.
  • compositions of the invention include an auristatin or an auristatin derivative or analog.
  • compositions of the invention contain monomethyl auristatin E (MMAE).
  • compositions of the invention contain monomethyl auristatin F (MMAF).
  • an immunoconjugate composition of the invention contains dolastatin or a dolastatin peptidic analog or derivative, e.g., an auristatin (see, e.g., U.S. Pat. Nos. 5,635,483, 5,780,588, and 5,663,149).
  • compositions of the invention include a maytansinoid molecule.
  • Maytansinoids are mitototic inhibitors which act by inhibiting tubulin polymerization. Methods of making maytansinoids and their therapeutic use are disclosed, for example, in U.S. Pat. Nos. 5,208,020; 5,416,064, 6,441,163 and European Pat. EP 0 425 235 B1; each of which is herein incorporated by reference in its entirety.
  • the cytotoxin is a maytansinoid or a maytansinoid derivative or analog.
  • Maytansinoid drug moieties are attractive drug moieties in antibody-drug conjugates because they are: (i) relatively accessible to prepare by fermentation or chemical modification or derivatization of fermentation products, (ii) amenable to derivatization with functional groups suitable for conjugation through non-disulfide linkers to antibodies, (iii) stable in plasma, and (iv) effective against a variety of tumor cell lines.
  • Maytansine compounds suitable for use as maytansinoid drug moieties are well known in the art, and can be isolated from natural sources according to known methods, produced using genetic engineering techniques (see Yu et al PNAS 99:7968-7973 (2002)), or maytansinol and maytansinol analogues can be prepared synthetically according to known methods.
  • compositions of the invention include the maytansinoid DM1 (N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine).
  • compositions of the invention include the maytansinoid DM2.
  • compositions of the invention include the maytansinoid DM3 (N(2′)-deacetyl-N-2-(4-mercapto-1-oxopentyl)-maytansine) or DM4 (N(2′)-deacetyl-N2-(4-mercapto-4-methyl-1-oxopentyl)-maytansine).
  • compositions of the invention include a cytoxic agent that is an alkylating agent.
  • the cytotoxic agent is selected from mechlorethamine, thiotepa, thioepa chlorambucil, melphalan, carmustine (BSNU), BCNU lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, and streptozoicin.
  • compositions of the invention include a cytoxic agent that is an antimetabolite.
  • the cytotoxic agent is selected from methotrexate, dichloromethotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil and 5-fluorouracil decarbazine.
  • the multivalent and multispecific composition-drug conjugate (e.g., MRD-containing antibody-drug conjugate) is capable of producing double-stranded DNA breaks.
  • the MRD-containing antibody-drug conjugate contains a member of the calicheamicin family of antibiotics capable of producing double-stranded DNA breaks at sub-picomolar concentrations.
  • a multivalent and multispecific composition-drug conjugate (e.g., MRD-containing antibody-drug conjugate) contains calicheamycin. For the preparation of conjugates of the calicheamicin family, see e.g., U.S. Pat. Nos.
  • Structural analogues of calicheamicin which can be contained in the multivalent and multispecific composition-drug conjugate (e.g., MRD-containing antibody-drug conjugate) of the invention include, but are not limited to, gamma 1 I , alpha 2 I , alpha 3 I , N-acetylamma 1 I , PSAG and theta 1 I (Hinman et al., Cancer Research 53:3336-3342 (1993), and Lode et al., Cancer Research 58:2925-2928 (1998).
  • multivalent and multispecific composition-drug conjugate e.g., MRD-containing antibody-drug conjugate
  • compositions of the invention include a cytoxic agent selected from adriamicin, doxorubicin, mitomycin C, busulfan, cytoxin, chlorambucil, etoposide, etoposide phosphate, CC-1065, duocarmycin, KW-2189, CC1065, taxotere (docetaxel), methopterin, aminopterin, topotecan, camptothecin, porfiromycin, bleomycin, teniposide, esperamicins, mithramycin, anthramycin (AMC), fludarabine, tamoxifen, taxotere (docetaxel), cytosine arabinoside (Ara-C), adenosine arabinoside, cisplatin, carboplatin, cis-dich
  • cytotoxins and chemotherapeutic agents are described in Remington's Pharmaceutical Sciences, 19th Ed. (Mack Publishing Co. 1995), and in Goodman and Gilman's The Pharmacological Basis of Therapeutics, 7th Ed. (MacMillan Publishing Co. 1985).
  • types of cytotoxins, linkers and other methods that can be use or routinely adapted to conjugate therapeutic agents to the MRD-comprising antibody complex, see e.g., Intl. Appl. Publ. WO2007/059404; Saito et al., Adv. Drug Deliv. Rev. 55:199-215 (2003); Trail et al., Cancer Immunol Immunother.
  • a multispecific and multivalent composition of the invention e.g., an MRD-containing antibody
  • Enzymatically active toxins and fragments thereof that can be used in compositions of the invention include, but are not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), Pseudomonas exotoxin, Pseudomonas endotoxin , ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, ribonuclease, DNase I, Staphylococcal enterotoxin-A, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes
  • peptide-based drug moieties can be prepared by forming a peptide bond between two or more amino acids and/or peptide fragments.
  • Such peptide bonds can be prepared, for example, according to the liquid phase synthesis method (see E. Schroder and K. Lubke, “The Peptides”, volume 1, pp. 76-136, 1965, Academic Press) that is well known in the field of peptide chemistry.
  • the auristatin/dolastatin drug moieties may be prepared according to the methods of: U.S. Pat. Nos. 5,635,483 and 5,780,588; Pettit et al., J. Am. Chem. Soc.
  • the compositions of the invention comprise a highly radioactive atom.
  • a variety of radioactive isotopes are available for the production of radioconjugated multivalent and multispecific compositions (e.g., MRD-containing antibodies). Examples include At 211 , I 131 , I 125 , Y. 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 to and radioactive isotopes of Lu.
  • the conjugate When used for detection, it may comprise a radioactive atom for scintiographic studies, for example tc 99m or I 123 , or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123 again, iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • the radio- or other labels can be incorporated in the conjugate using techniques known in the art.
  • the peptide can be biosynthesized or can be synthesized by chemical amino acid synthesis using suitable amino acid precursors involving, for example, fluorine-19 in place of hydrogen.
  • Labels such as tc 99m or I 123 , Re 186 , Re 188 and In 111 can be attached via a cysteine residue in the peptide.
  • Yttrium-90 can be attached via a lysine residue.
  • the IODOGEN method (Fraker et al Biochem. Biophys. Res. Commun. 80: 49-57 (1978)) can be used to incorporate iodine-123. “Monoclonal Antibodies in Immunoscintigraphy” (Chatal, CRC Press 1989) describes in detail other methods that can be routinely applied to label the compositions of the invention.
  • a linker can be a “cleavable linker,” facilitating release of a drug in the cell.
  • an acid-labile linker e.g., hydrazone
  • protease-sensitive linker e.g., peptidase-sensitive
  • photolabile linker dimethyl linker or disulfide-containing linker
  • the invention encompasses multivalent and multispecific compositions containing one or more linkers that can contain any of a variety of groups as part of its chain that will cleave in vivo, e.g., in a cell, at a rate which is enhanced relative to that of constructs that lack such groups.
  • conjugates of the linker arms with therapeutic and diagnostic agents are useful to form prodrug analogs of therapeutic agents and to reversibly link a therapeutic or diagnostic agent (e.g., a cytotoxin or MRD) to a targeting agent, a detectable label, or a solid support.
  • the linkers can be stable in plasma so as not to release an MRD or cytotoxic agent. In the case of cytotoxins the linkers can be stable in plasma and labile once internalized so as to release the cytotoxin in an active form.
  • MRDs and/or cytotoxic agents are optionally attached to one another or to the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) of the invention with a linker as described herein or otherwise known in the art.
  • Conjugates of the MRD-containing antibody with an MRD or a cytotoxic agent can be made using a variety of bifunctional protein coupling agents known in the art, including, but not limited to, coupling agents containing a group selected from 6-maleimidocaproyl (MC), maleimidocaproyl-polyethylene glycol (“MC(PEG) 6 -OH” (amenable to attachment to antibody cysteines)), maleimidopropanoyl (MP), MPBH, valine-citrulline (val-cit (exemplary dipeptide in a protease cleavable linker)), methyl-valine-citrulline (“Me-Val-CitN,” a linker in which a peptide bond has been modified to prevent its
  • the multivalent and monovalent multispecific composition is covalently attached to a cytotoxic agent, via a linker at 1-5, 5-10, 1-10, or 1-20 sites on the multivalent and multispecific composition.
  • the multivalent and monovalent multispecific composition is covalently attached to a cytotoxic agent via a linker at more than 2, 5 or 10 sites on the multivalent and multispecific composition.
  • the multivalent and monovalent multispecific composition (e.g., MRD containing antibody) complex is associated with a prodrug.
  • a prodrug e.g., MRD containing antibody
  • Prodrug synthesis, chemical linkage to antibodies, and pharmacodynamic properties are known in the art and can routinely be applied to make and use multivalent and multivalent compositions of the invention that contain prodrugs, such as, MRD-containing antibody-prodrug compositions. See, e.g., Intl Publ. No. WO96/05863 and in U.S. Pat. No. 5,962,216, each of which is herein incorporated by reference in its entirety.
  • a fusion protein comprising an antibody and a cytotoxic agent can be made, e.g., by recombinant techniques or peptide synthesis.
  • a recombinant DNA molecule can comprise regions encoding the antibody and cytotoxic portions of the conjugate either adjacent to one another or separated by a region encoding a linker peptide which does not destroy the desired properties of the conjugate.
  • the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) composition of the invention also can be conjugated to a radioactive isotope to generate cytotoxic radiopharmaceuticals, also referred to as radioimmunoconjugates.
  • radioactive isotopes that can be conjugated to multivalent and monovalent multispecific compositions (e.g., MRD containing antibodies) for use diagnostically or therapeutically include, but are not limited to, iodine 131 , indium 111 , yttrium 90 , and lutetium 177 . Methods for preparing radioimmunconjugates are established in the art.
  • radioimmunoconjugates examples include ZevalinTM (IDEC Pharmaceuticals) and BexxarTM (Corixa Pharmaceuticals), and similar methods can be used to prepare radioimmunoconjugates using the MRD-containing antibodies of the invention.
  • linker-drug moieties to cell-targeted proteins such as antibodies are known in the art and include those described for example, in U.S. Pat. Nos. 5,208,020 and 6,441,163; Intl. Appl. Publ. Nos. WO2005037992, WO2005081711, and WO2006/034488, each of which is herein incorporated by reference in its entirety. See, also e.g., Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy”, in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al., (eds.), pp. 243-56 (Alan R.
  • a multivalent and monovalent multispecific composition of the invention comprising a cytotoxic agent (e.g., an MRD-containing antibody-cytotoxic agent conjugate) and may generally be referred to herein as an immunoconjugate.
  • an immunoconjugate of the invention binds a cell surface target that is internalized into the cell.
  • the binding of an immunoconjugate of the invention (e.g., an MRD-containing antibody-cytotoxic agent conjugate) to a cell surface target results in the internalization of the immunoconjugate into the cell in vitro.
  • the binding of immunoconjugate to a cell surface target results in the internalization of the composition into the cell in vivo.
  • Methods for treating a patient described herein can comprise: administering to the patient a therapeutically effective amount of an immunoconjugate (e.g., a multivalent and monovalent multispecific composition of the invention comprising a cytotoxic agent, such as an MRD-containing antibody-cytotoxic agent conjugate) that comprises a cytotoxic agent and binds a target that is internalized into a cell.
  • an immunoconjugate e.g., a multivalent and monovalent multispecific composition of the invention comprising a cytotoxic agent, such as an MRD-containing antibody-cytotoxic agent conjugate
  • the immunoconjugate comprises a cytotoxic agent disclosed herein.
  • the immunoconjugate comprises a cytotoxic agent selected from an alkylating agent, antiproliferative agent, tubulin binding agent, vinca alkaloid, enediyne, podophyllotoxin, podophyllotoxin derivative, a member of the pteridine family of drugs, taxane, a dolastatin, topoiosomerase inhibitor, or a platinum complex chemotherapeutic agent.
  • the cytoxic agent is a maytansinoid or a maytansinoid derivative or analog.
  • the cytoxic agent is the maytansinoid DM1, DM2, or DM3.
  • the cytotoxic agent is auristatin or an auristatin derivative or analog.
  • the cytoxic agent is MMAE or MMAF.
  • the cytotoxic agents are optionally attached to the other components of the immunoconjugate by a linker.
  • the cytotoxic agent is attached to the other components of the immunoconjugate by an enzyme cleavable linker.
  • the cytotoxic agent is attached to the other components of the immunoconjugate by an acid-labile linker.
  • the cytoxic agent of an immunoconjugate of the invention has a free drug potency of less than 10 ⁇ 7 M, 10 ⁇ 8 M, or 10 ⁇ 9 M.
  • the cytoxin has a free drug potency of 10 ⁇ 8 to 10 ⁇ 11 M.
  • a target bound by the immunoconjugate is selected from CD19, CD22, CD30, CD33, CD56, CD70, CD79a, CD80, CD83, CD95, CD126, CD133, CD138, PSMA, EphA2, ErbB2 (CD340), SLC44A4, MN (carbonic anhydrase IX), GPNMB (glycoprotein non-metastatic melanoma protein), Cripto, and ⁇ V integrin.
  • a target bound by the immunoconjugate is selected from CD1, CD1a, CD2, CD3, CD4, CD5, CD8, CD11A, CD14, CD15, CD16, CL18, CD19, CD20, CD25, TNFRSF5 (CD40), CD64, CD74, CD79, CD105, CD174, CD205, CD227, CD326, CD340, MUC16, EGP-1, EGP-2, EGF receptor (ErbB1), ErbB2, ErbB3, Factor H, FHL-1, Flt-3, folate receptor, Ga 733, GROB, HMGB-1, hypoxia inducible factor (HIF), HM1.24, HER-2/neu, insulin-like growth factor (ILGF), IFN-gamma, IFN-alpha, IFN-beta, IL2R, IL4R, IL6R, IL13R, IL15R, IL17R, IL18R, IL2, IL6, IL8, IL12, IL15,
  • a target bound by the immunoconjugate is a myeloid and hematopoietic target selected from CD33, CD64, TNFRSF5 (CD40), CD56, and CD138.
  • a target bound by the immunoconjugate is a carcinoma target selected from EpCam, GD2, EGFR, CD74, CD227, CD340, MUC16, GD2, GPNMB, PSMA, crypto, TMEFF2, EphB2, 5t4, mesothelin, TAG-72, and MN.
  • a target bound by the immunoconjugate is a B cell target selected from CD19/CD21, CD20, CD22, TNFRSF5 (CD40), CD70, CD79a, CD79b, and CD205.
  • a target bound by the immunoconjugate is a T cell target selected from CD25, CD30, TNFRSF5 (CD40), CD70, and CD205.
  • a target bound by an endothelial cell target selected from CD105, the stromal cell target FAP, and the vascular target ED-B.
  • a fusion protein comprising the antibody and cytotoxic agent may be made, e.g., by recombinant techniques or peptide synthesis.
  • the length of DNA may comprise respective regions encoding the two portions of the conjugate either adjacent one another or separated by a region encoding a linker peptide which does not destroy the desired properties of the conjugate.
  • an immunoconjugate has in vitro or in vivo cell killing activity.
  • the linker is attached to the antibody through a thiol group on the antibody.
  • the linker is cleavable by a protease.
  • the linker comprises a val-cit dipeptide.
  • the linker comprises a p-aminobenzyl unit.
  • the p-aminobenzyl unit is disposed between the drug and a protease cleavage site in the linker.
  • the p-aminobenzyl unit is p-aminobenzyloxycarbonyl (PAB).
  • the linker comprises 6-maleimidocaproyl.
  • the 6-maleimidocaproyl is disposed between the antibody and a protease cleavage site in the linker. The above embodiments may occur singly or in any combination with one another.
  • the MRD-containing antibody of the present invention may also be conjugating to a prodrug-activating enzyme which converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see e.g., WO81/01145) to an active anti-cancer drug.
  • a prodrug e.g., a peptidyl chemotherapeutic agent, see e.g., WO81/01145
  • the enzyme component of the immunoconjugate is preferably capable of acting on a prodrug in such a way so as to convert it into its more active, cytotoxic form.
  • Enzymatically active toxins and fragments thereof which can be used include diphtheria A chain, non-binding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, eromycin and the tricothecenes. See, for example, WO93/21232.
  • the multivalent and multispecific compositions of the invention are conjugated to a radioisotope, such as, 90 Y, 125 I, 131 I, 123 I, 111 In, 105 Rh, 153 Sm, 67 Cu, 67 Ga, 166 Ho, 177 Lu, 186 Re and 188 Re using anyone of a number of well-known chelators or direct labeling.
  • the MRD-containing antibody is coupled to drugs, prodrugs or lymphokines such as, interferon.
  • compositions of the invention can be labeled with ligand reagents that bind, chelate or otherwise complex a radioisotope metal where the reagent is reactive with the engineered cysteine thiol of the antibody, using techniques known in the art such as, those described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley-Interscience, New York, N.Y. Pubs. (1991).
  • Chelating ligands which may complex a metal ion and that may have use in the compositions and methods of the invention include DOTA, DOTP, DOTMA, DTPA and TETA (Macrocyclics, Dallas, Tex.).
  • Radionuclides can be targeted via complexation with the antibody-drug conjugates of the invention (Wu et al Nature Biotechnology 23(9): 1137-1146 (2005)).
  • Linker reagents such as, DOTA-maleimide (4-maleimidobutyramidobenzyl-DOTA) can be prepared by the reaction of aminobenzyl-DOTA with 4-maleimidobutyric acid (Fluka) activated with isopropylchloroformate (Aldrich), following the procedure of Axworthy et al., Proc. Natl. Acad. Sci. USA 97(4):1802-1807 (2000)).
  • DOTA-maleimide reagents react with the free cysteine amino acids of the cysteine engineered antibodies and provide a metal complexing ligand on the antibody (Lewis et al., Bioconj. Chem. 9:72-86 (1998)).
  • Chelating linker labeling reagents such as, DOTA-NHS (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid mono (N-hydroxysuccinimide ester) are commercially available (Macrocyclics, Dallas, Tex.).
  • Conjugates of the multivalent and multispecific compositions of the invention can routinely be made using a variety of bifunctional protein-coupling agents such as, N-succinimidyl-3-(2-pyridyidithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as, dimethyl adipimidate HCL), active esters (such as, disuccinimidyl suberate), aldehydes (such as, glutareldehyde), bis-azido compounds (such as, bis(p-azidobenzoyl)hexanediamine), bis-diazonium derivatives (such as, bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as, tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as, 1,5-d
  • SPDP N-succinimidyl-3-(2-
  • the toxin is conjugate to an MRD-containing antibody through an enzyme-cleavable linker system (e.g., such as, that present in SGN-35).
  • an enzyme-cleavable linker system e.g., such as, that present in SGN-35.
  • Conjugates of an MRD-containing antibody and one or more small molecule toxins such as, a calicheamicin, maytansinoids, a trichothene, and CC 1065, and the derivatives of these toxins that have toxin activity, can also be used.
  • the MRD-containing antibody can be complexed, or have MRDs that bind with other immunologically active ligands (e.g., chemokines, cytokines, and antibodies or fragments thereof) wherein the resulting molecule binds to the neoplastic cell or other target as well as the chemokine, cytokine, or an effector cell such as, a T cell.
  • these conjugates can be generated as fusion proteins.
  • the enzymes of this invention can be covalently bound to the antibody by techniques well-known in the art such as, the use of the heterobifunctional crosslinking reagents discussed above.
  • fusion proteins comprising at least the antigen-binding region of an antibody of the invention linked to at least a functionally active portion of an enzyme of the invention can be constructed using recombinant DNA techniques known in the art.
  • the N-terminus or C-terminus of the antibody to which an MRD is operably linked in the MRD-antibody fusions is truncated. In preferred embodiments, this truncation does not prevent or reduce the ability of the antibody to bind to its target antigen via its antigen binding domain. In other embodiments, the truncation does not prevent or reduce Fc effector function, half-life and/or ADCC activity.
  • MRDs are attached in the terminal region of the antibody chain. More particularly, in certain embodiments, the MRD is attached within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 residues of the C-terminal amino acid of the heavy chain.
  • the MRD is attached within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 residues of the C-terminal amino acid of the light chain. In additional embodiments, the MRD is attached within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 residues of the N-terminal amino acid of the heavy chain. In other embodiments, the MRD is attached within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 residues of the N-terminal amino acid of the light chain.
  • a MRD that is linked to the N-terminal end of the heavy chain can be linked to the first, second, third, fourth, fifth, or tenth amino acid or the N-terminal chain of the heavy chain.
  • an MRD-antibody fusion containing an MRD linked to the N-terminal of the heavy chain may contain amino acids 1-3 of the heavy chain sequence linked to the MRD, which is linked to amino acid 4 of the heavy chain sequence.
  • one or more MRDs are attached to an antibody at locations other than the termini of the antibody light and heavy chains.
  • the MRD can be attached to any portion of the antibody that does not prevent the ability of the antibody to bind its target.
  • the MRD is located outside the antibody combining site.
  • the MRD can be located within a heavy chain sequence or within a light chain sequence.
  • the MRD can be located between the Fc domain and the hinge region, between the hinge region and the CH1 domain of the heavy chain, between the CH1 domain and the variable region of the heavy chain, or between the constant region and the variable region of the light chain.
  • Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathways have been observed to provide at best transitory therapeutic benefits followed by restoration of tumor growth and progression due to an apparent ability of angiogenic tumors to adapt to the presence of these inhibitors.
  • VEGF vascular endothelial growth factor
  • an MRD-containing antibody binds 2 or more targets selected from: VEGF (i.e., VEGFA), VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TIE2, TNFSF2 (TNFa), FGFR3, EFNa1, EFNa2, ANG1, ANG2, IL6, IL8, IL18, HGF, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR2, VEGFR3, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ v ⁇ 3 integrin, TGFb, M
  • Multivalent and multispecific compositions that bind VEGF and 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGF.
  • the antibody component of the MRD-containing antibody is bevacizumab.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind VEGF and 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGF.
  • the antibody component of the MRD-containing antibody is bevacizumab.
  • an MRD-containing antibody binds VEGF (i.e., VEGFA) and additionally binds an angiogenic target selected from: VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, EFNa1, EFNa2, ANG1, ANG2, IL-6, IL-8, IL-18, HGF, TIE2, PDGFA, P1GF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR2, VEGFR3, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ v ⁇ 3
  • Multivalent and multispecific compositions that bind VEGF and 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGF.
  • the antibody component of the MRD-containing antibody is bevacizumab.
  • the antibody component of the MRD-containing antibody competes for VEGF binding with bevacizumab.
  • an MRD-containing antibody binds TNF alpha and additionally binds a target selected from: Te38, IL-12, IL-12p40, IL-13, IL-15, IL-17, IL-18, IL-1beta, IL-23, MIF, PEG2, PGE4, VEGF, TNFSF11 (RANKL), TNFSF13B (BLYS), GP130, CD-22, and CTLA-4.
  • an MRD-containing antibody binds TNF alpha, IL6, and TNFSF13B (BLYS).
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind TNF and 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TNF.
  • the antibody component of the MRD-containing antibody is adalimumab, certolizumab, golimumab or AME-527.
  • the antibody component of the MRD-containing antibody competes for TNF binding with adalimumab, certolizumab, golimumab or AME-527.
  • an MRD-containing antibody binds IL1 alpha and IL1 beta. In another embodiment, an MRD-containing antibody binds IL1 beta and TNFSF11 (RANKL). In an additional embodiment, an MRD-containing antibody binds IL1 beta and a target selected from IL13, IL17A, TNF, VEGF, PGE2, VEGFR1, VEGFR2, TNFSF12 (TWEAK) and TNF. Multivalent and multispecific compositions (e.g., MRD-containing antibodies) that bind IL1 beta and at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention. In specific embodiments, the antibody component of the MRD-containing antibody binds IL 1 beta.
  • the antibody component of the MRD-containing antibody is catumaxomab, Xoma052, canakinumab or ACZ885. In additional embodiments, the antibody component of the MRD-containing antibody competes for IL1 alpha or IL1 beta binding with catumaxomab, Xoma052, canakinumab or ACZ885.
  • an MRD-containing antibody binds IL12.
  • an MRD-containing antibody binds IL 12 and additionally binds IL18 or TNFSF12 (TWEAK).
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds CTLA-4.
  • the antibody component of the MRD-containing antibody is briakinumab or ustekinumab.
  • the antibody component of the MRD-containing antibody competes for IL12 binding with briakinumab or ustekinumab.
  • an MRD-containing antibody binds CTLA-4.
  • an MRD-containing antibody binds CTLA4 and additionally binds PDL-1 or BTNO2.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds CTLA-4.
  • the antibody component of the MRD-containing antibody is tremelimumab or iplimumab.
  • the antibody component of the MRD-containing antibody competes for CTLA-4 binding with tremelimumab or iplimumab.
  • an MRD-containing binds IL13.
  • an MRD-containing antibody binds IL13 and additionally binds a target selected from: IL1beta, IL4, IL9, IL13, IL25, a LHR agonist, MDC, MIF, PED2, SPRR2a, SPRR2b; TARC, TGF-beta and IL25.
  • an MRD-containing antibody binds IL13 and a target selected from IL5, ADAM8, a LHR (agonist), IL23p19 and IgE.
  • Multivalent and multispecific compositions that bind IL13 and at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds IL13.
  • the antibody component of the MRD-containing antibody is TNX-650, lebrikizumab or CAT354.
  • the antibody component of the MRD-containing antibody competes for IL13 binding with TNX-650, lebrikizumab or CAT354.
  • an MRD-containing antibody binds RGM A.
  • an MRD-containing antibody binds RGM A and additionally binds a target selected from RGM B, MAG, NgR, NogoA, OMGp and CSPGs.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds RGM A.
  • an MRD-containing antibody binds CD38 and additionally binds a target selected from CD20, TNFRSF5 (CD40) ALK1, TNF, VEGF, VEGFA, VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, VEGFR1, VEGFR2 and CD138.
  • Multivalent and multispecific compositions that bind CD38 and at least 1, 2 or all 3 of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD38.
  • the antibody component of the MRD-containing antibody binds MOR202 or daratumumab.
  • the antibody component of the MRD-containing antibody competes for CD38 binding with MOR202 or daratumumab.
  • an MRD-containing antibody binds ErbB1 (EGFR) and additionally binds ErbB3.
  • the antibody component of the MRD-containing antibody binds ErbB1.
  • the antibody component of the MRD-containing antibody is ERBITUX®.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for ErbB1-binding with ERBITUX®.
  • the antibody component of the MRD-containing antibody is an ErbB1-binding antibody selected from: nimotuzumab, zalutumumab, matuzumab, panitumumab, MEDX-214, and ABX-EGF.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for ErbB1-binding with an antibody selected from: nimotuzumab, zalutumumab, matuzumab, panitumumab, MEDX-214, and ABX-EGF.
  • an MRD-containing antibody binds ErbB2 and IGF1R. In another embodiment, an MRD-containing antibody binds ErbB2, Ang2, and IGF1R. In specific embodiments, the antibody component of the MRD-containing antibody binds ErbB2. In additional embodiments, the antibody component of the MRD-containing antibody is HuMax-Her2TM or trastuzumab-DM1. In further embodiments, the antibody component of the MRD-containing antibody is trastuzumab. In additional embodiments, the antibody component, MRD component, and/or MRD-containing antibody competes for ErbB2-binding with trastuzumab.
  • an MRD-containing antibody binds ErbB2 and additionally binds a target selected from: ErbB3, EGFR, IGF1R, cMet, VEGF, RON (MST1R), DLL4, PLGF, CDCP1 (CD318), NRP1, TNFRSF10A (DR4) and TNFRSF10B (DR5).
  • an MRD-containing antibody binds ErbB2 and additionally binds, a target selected from: CD2, CD3, CD4 and NKG2D.
  • an MRD-containing antibody binds ErbB2 and IGF1, IGF2 or IGF1,2.
  • Multivalent and multispecific compositions that bind ErbB2 and additionally bind 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds ErbB2.
  • the antibody component of the MRD-containing antibody is HuMax-Her2TM or trastuzumab-DM1.
  • the antibody component of the MRD-containing antibody is trastuzumab.
  • the antibody component, MRD component, and/or MRI)-containing antibody competes for ErbB2-binding with trastuzumab.
  • an MRD-containing antibody binds ErbB2 and additionally binds ErbB3.
  • the antibody component of the MRD-containing antibody binds ErbB2.
  • the antibody component of the MRD-containing antibody is HuMax-Her2TM trastuzumab-DM1.
  • the antibody component of the MRD-containing antibody is trastuzumab.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for ErbB2-binding with trastuzumab.
  • the antibody component of the MRD-containing antibody is an ErbB2-binding antibody selected from: MDX-210 (Medarex), tgDCC-E1A (Targeted Genetics), MGAH22 (MacroGenics), and pertuzumab (OMNITARGTM).
  • the antibody component, MRD component, and/or MRD-containing antibody competes for ErbB2-binding with an antibody selected from: MDX-210, tgDCC-E1A, MGAH22, and pertuzumab.
  • an MRD-containing antibody binds ErbB2 and HER2/3. In further embodiments, an MRD-containing antibody binds ErbB2 and HER2/3 simultaneously.
  • Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathways have been observed to provide at best transitory therapeutic benefits followed by restoration of tumor growth and progression due to an apparent ability of angiogenic tumors to adapt the presence of these inhibitors.
  • VEGF vascular endothelial growth factor
  • an MRD-containing antibody binds PDGFRA and additionally binds an target selected from: VEGFA, VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGRF1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, EFNa1, EFNa2, ANG1, ANG2, 1L6, IL8, IL18, IGF1, IGF2, IGF1,2, HGF, TIE2, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR2, VEGFR3, EGFR, cMET, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integr
  • Multivalent and multispecific compositions that bind PDGFRA and binds at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds PDGFRA.
  • the antibody component of the MRD-containing antibody is olaratumab.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for PDGFRA binding with olaratumab.
  • the antibody component of the MRD-containing antibody is MEDI-575.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for PDGFRA binding with MEDI-575.
  • an MRD-containing antibody binds PDGFRB and additionally binds an target selected from: VEGFA, VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, EFNa1, EFNa2, ANG1, ANG2, IL6, IL8, IL18, IGF1, IGF2, IGF1,2, HGF, TIE2, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR2, VEGFR3, EGFR, cMET, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integr
  • Multivalent and multispecific compositions that bind PDGFRB and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds PDGFRB.
  • an MRD-containing antibody binds VEGFR1 and additionally binds an angiogenic target selected from: VEGF (i.e., VEGFA), VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, EFNa1, EFNa2, ANG1, ANG2, IL6, IL8, IL18, HGF, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR2, VEGFR3, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ v ⁇ 3 integrin, T
  • Multivalent and multispecific compositions that bind VEGFR1 and additionally bind 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGFR1.
  • the antibody component of the MRD-containing antibody is IMC-18F1.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for VEGFR1 binding with IMC-18F1.
  • an MRD-containing antibody hinds VEGFR2 and additionally binds a target selected from: VEGF (i.e., VEGFA), VEGFB, FGF1, FGF2, FGF4, FGF7, FGF8b, FGF19, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIc), FGFR3, TNFSF2 (TNFa), FGFR3, NRP1, ROBO4, CD30, CD33, CD55 CD80, KIT, CXCL12, Notch1EFNa1, EFNa2, ANG1, ANG2, IL6, IL8, IL18, HGF, PDGFA, PLGF, PDGFB, CXCL12, KIT, GCSF, CXCR4, PTPRC, TIE2, VEGFR1, VEGFR3, Notch 1, DLL4, EGFL7, ⁇ 2 ⁇ 1 integrin, ⁇ 4
  • Multivalent and multispecific compositions that bind VEGFR2 and additionally bind 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGFR2.
  • the antibody component of the MRD-containing antibody is IMC-1C11 or DC101.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for VEGFR2 binding with IMC-1C11 or DC101.
  • an MRD-containing antibody binds VEGFR2 and additionally binds ANG2 or TIE2.
  • the antibody component of the MRD-containing antibody binds VEGFR2.
  • the antibody component of the MRD-containing antibody is IMC-1C11, DC101 or TTAC-0001.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for VEGFR2 binding with IMC-1C11, DC101 or TTAC-0001.
  • the TIE2 binding component comprises a fragment of ANG2 that binds TIE2.
  • the TIE2 binding component comprises amino acids 283-449 of the human ANG2 disclosed in NCBI Ref. Seq. No. NP — 001138.1.
  • an MRD-containing antibody binds DLL4 and additionally binds a target selected from: EGFR, PLGF, VEGFR1, VEGFR2 and VEGF.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody is REGN421.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for DLL4 binding with REGN421.
  • an MRD-containing antibody binds to an anti-angiogenic and a metastatic or invasive cancer target.
  • an MRD-containing antibody binds to an angiogenic target and also binds a metastatic or invasive cancer target selected from: CXCL12, CXCR4 (e.g., CXCR4b), CCR7 (e.g., CXCR7b), CD44 (e.g., CD44v3 and CD44v6), ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 5 ⁇ 31 integrin, ⁇ v ⁇ 1 integrin, ⁇ v ⁇ 3 integrin, TGFb, ⁇ v ⁇ 5 integrin, ⁇ 9 ⁇ 31 integrin, ⁇ 6 ⁇ 4 integrin, ⁇ M ⁇ 2 integrin, PD-1, HGF, cMET, MMP2, MMP-7, MMP-9, MMP-12, VEGFA, VEGFB, and IGF1.
  • Multivalent and multispecific compositions that bind an angiogenic target and also bind 2, 3, 4, 5 or more of these metastatic or invasive cancer targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds VEGF.
  • the antibody component of the MRD-containing antibody is bevacizumab.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for VEGF binding with bevacizumab.
  • an MRD-containing antibody binds to 2 or more targets associated with distinct cell signaling pathways. In additional embodiments, an MRD-containing antibody binds to 2 or more targets associated with redundant, overlapping or cross-talking signaling pathways. For example, in one embodiment, an MRD-containing antibody binds to 2 or more targets associated with PI3K/AKT/mTOR signaling (e.g., ErbB2, EGFR, IGF1R, Notch, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIb), FGFR3, FGFR4, GPCR, and/or c-MET). In some embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) binds 2, 3, 4, 5 or more of these targets.
  • PI3K/AKT/mTOR signaling e.g., ErbB2, EGFR, IGF1R,
  • an MRD-containing antibody binds to 2 or more targets associated with receptor tyrosine Raf/MEK/MAPK signaling (e.g., VEGFR1, VEGFR2, VEGFR3, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIb), FGFR3, FGFR4, CD28, RET, cMET, EGFR, ErbB2, Notch, Notch1, Notch3, Notch4, DLL1, DLL4, Jagged, Jagged1, Jagged2, and Jagged3.
  • the multivalent and multispecific compositions bind 1, 2, 3, 4, 5 or more of these targets.
  • an MRD-containing antibody binds to 2 or more targets associated with SMAD signaling (e.g., Notch, TGF ⁇ , TGF ⁇ R1, TGF ⁇ R2, and a BMP).
  • targets associated with SMAD signaling e.g., Notch, TGF ⁇ , TGF ⁇ R1, TGF ⁇ R2, and a BMP.
  • the multivalent and multispecific compositions bind 2, 3, 4, 5 or more of these targets.
  • an MRD-containing antibody binds to 2 or more targets associated with JAK/STAT signaling (e.g., IFNgR1, IFNgR3, IFNG, IFN-AR2, IFN-AR1, IFN alpha, IFN beta, IL6a receptor (GP130), IL6, IL12R131, IL12, and EGFR).
  • targets associated with JAK/STAT signaling e.g., IFNgR1, IFNgR3, IFNG, IFN-AR2, IFN-AR1, IFN alpha, IFN beta, IL6a receptor (GP130), IL6, IL12R131, IL12, and EGFR.
  • the invention encompasses an MRD-containing antibody that binds to 2 or more targets selected from WNT1, WNT2, WNT2b, WNT3, WNT3A, WNT4, WNT5A, WNT5B, WNT6, WNT7A, WNT7B, WNT8A, WNT8B, WNT9A, WNT9B, WNT10A, WNT1013, WNT11, WNT16, FZD1, FZD2, FZD4, FZD5, FZD6, FZD7, FZD8, Notch, Notch1, Notch3, Notch4, DLL1, DLL4, Jagged, Jagged1, Jagged2, and Jagged3.
  • the multivalent and multispecific compositions bind 2, 3, 4, 5 or more of these targets.
  • an MRD-containing antibody binds to 2 or more targets associated with NFkB signaling (e.g., BCR, TCR, IL1R, IL1, FZD1, FZD2, FZD4, FZD5, FZD6, FZD7, FZD8, Notch, Notch1, Notch3, Notch4, DLL4, Jagged, Jagged1, Jagged2, Jagged3, TNFSF1 (TNFb, LTa), TNFRSF1A (TNFR1, p55, p60), TNFRSF1B (TNFR2), TNFSF6 (Fas LigaLd), TNFRSF6 (Fas, CD95), TNFRSF6B (DcR3), TNFSF7 (CD27 Ligand, CD70), TNFRSF7 (CD27), TNFSF8 (CD30 Ligand), TNFRSF8 (CD30), TNFSF11 (RANKL), TNFRSF11A (RANK), TNFSF12 (TWEAK), TNFRSF12
  • an MRD-containing antibody binds to 2 or more targets associated with cell proliferation (e.g., FGF1, FGF2, FGF7, FGF4, FGF10, FGF18b, FGF19, FGF23, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFRIIIB and FGFR-IIIC), FGFR3, FGFR4, TCR, TNFRSF5 (CD40), TLR1, TLR2, TLR3, TLR 4, TLR5, and TLR6).
  • the multivalent and multispecific compositions bind 2, 3, 4, 5 or more of these targets.
  • an MRD-containing antibody binds to 2 or more targets associated with toll-like receptor signaling (e.g., TLR1, TLR2, TLR3, TLR 4, TLR5, and TLR6).
  • targets associated with toll-like receptor signaling e.g., TLR1, TLR2, TLR3, TLR 4, TLR5, and TLR6.
  • an MRD-containing antibody binds to 2 or more targets associated with B cell signaling (e.g., mIg, Ig ⁇ /Ig ⁇ (CD79a/CD79b) heterodimers ( ⁇ / ⁇ ), CD19, CD20, CD21, CD22, CD23, CD27, CD30, CD46, CD80, CD86, ICOSL (B7-H2), HLA-DR (CD74), PD1, PDL1, TNFRSF1A (TNFR1, p55, p60), TNFRSF1B (TNFR2), TNFRSF13B (TACI), TNFRSF13C (BAFFR), TNFRSF17 (BCMA), BTLA, TNFRSF5 (CD40), TLR4, TNFRSF14 (HVEM), Fc gamma RIIB, IL4R and CRAC.
  • B cell signaling e.g., mIg, Ig ⁇ /Ig ⁇ (CD79a/CD79b) heterodimers ( ⁇ /
  • the MRD-containing antibody binds to CD19 and CD20. In an additional embodiment, the MRD-containing antibody binds CD19, CD20, and CD22. In some embodiments, the multivalent and monovalent multispecific composition (e.g., MRD-containing antibodies) binds 2, 3, 4, 5 or more of these targets.
  • an MRD-containing antibody binds to 1 or more B cell surface markers selected from: CD10, CD24, CD37, CD53, CD72, CD75, CD77, CD79a, CD79b, CD81, CD82, CD83, CD84 (SLAMS) and CD85.
  • an MRD-containing antibody binds to 1 or more B cell surface markers selected from: CD 10, CD24, CD37, CD53, CD72, CD75, CD77, CD79a, CD79b, CD81, CD82, CD83, CD84 (SLAMS) and CD85.
  • the multivalent and multispecific compositions bind 2, 3, 4, 5 or more of these B cell surface markers.
  • an MRD-containing antibody binds CD19 and a target
  • an MRD-containing antibody binds CD20 and a target selected from: CD3, CD4 and NKG2D. Multivalent and multispecific compositions (e.g., MRD-containing antibodies) that bind CD19 and also bind at least 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD19.
  • the antibody component of the MRD-containing antibody is MDX-1342, SGN-CD19A, XMAB®5574, SGN-19A, ASG-5ME or MEDI-551.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for CD19 binding with MDX-1342, SGN-CD19A, XMAB®5574, SGN-19A, ASG-5ME or MEDI-551.
  • an MRD-containing antibody binds CD22 and a target selected from: CD19, CD20, CD23, CD30, CD33, TNFRSF5 (CD40), CD52, CD74, CD80, TNFRSF10A (DR4), TNFRSF10B (DR5), VEGF, TNF and NGF.
  • an MRD-containing antibody binds CD22 and a target selected from: CD3, CD4 and NKG2D.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind CD22 and also bind 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD22.
  • the antibody component of the MRD-containing antibody is epratuzumab or inotuzumab. In additional embodiments, the antibody component, MRD component, and/or MRD-containing antibody competes for CD22 binding with epratuzumab or inotuzumab.
  • the antibody component of the MRD-containing antibody is moxetumomab (CAT-8015, Cambridge Antibody Technologies). In additional embodiments, the antibody component, MRD component, and/or MRD-containing antibody competes for CD22 binding with moxetumomab.
  • an MRD-containing antibody binds TNFRSF5 (CD40) and a target selected from: BCMA, TNFSF11 (RANKL), VEGFR1, VEGFR2, TNFRSF10A (DR4), TNFRSF10B (DR5), CD22, CD30, CD38, CD56 (NCAM), CD70, CD80, CD138, IL6, IGF1, IGF2, IGF1,2, BLyS, APRIL and NGF.
  • an MRD-containing antibody binds CD40 and a target selected from: CD3, CD4 and NKG2D.
  • Multivalent and multispecific compositions that bind CD40 and also bind 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD40.
  • the antibody component of the MRD-containing antibody is CP870893, dacetuzumab, ANTOVA®, lucatumumab, XMAB®5485 or teneliximab.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for CD40 binding with CP870893, dacetuzumab, ANTOVA®, lucatumumab, XMAB®5485 or teneliximab.
  • an MRD-containing antibody binds CD33 and a target selected from: FLT3, CD44, TNFRSF10A (DR4), TNFRSF10B (DR5), CD80, MGC, VEGFR1, VEGFR2, IL1, IL6, TNF and VEGF.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the antibody component of the MRD-containing antibody binds CD33.
  • the antibody component of the MRD-containing antibody is gemtuzumab or lintuzumab.
  • the antibody component, MRD component, and/or MRD-containing antibody competes for CD33 binding with gemtuzumab or lintuzumab.
  • an MRD-containing antibody binds to 2 or more targets associated with antigen presentation cell signaling (e.g., mIg, Ig ⁇ /Ig ⁇ (CD79a/CD79b) heterodimers ( ⁇ / ⁇ ), CD19, CD20, CD21, CD22, CD23, CD27, CD28, CD30, CD30L, TNFSF14 (LIGHT, HVEM Ligand), CD70, ICOS, ICOSL (B7-H2), CTLA4, PD-1, PDL1 (B7-H1), B7-H4, B7-H3, PDL2 (B7-DC), BTLA, CD46, CD80 (B7-1), CD86 (B7-2), HLA-DR, CD74, PD1, TNFRSF4 (OX40), TNFRSF9 (41BB), TNFSF4 (OX40 Ligand), TNFSF9 (41BB Ligand), TNFRSF9 (41BB), TNFRSF1A (TNFR1, p55, p60
  • an MRD-containing antibody binds to 2 or more targets associated with T cell receptor signaling (e.g., CD3, CD4, CD27, CD28, CD70, IL2R, LFA-1, C4, ICOS, CTLA-4, CD45, CD80, CD86, PG-1, TIM1, TIM2, TIM3, TIM4, galectin 9, TNFRSF1A (TNFR1, p55, p60), TNFRSF1B (TNFR2), TNFRSF21 (DR6), TNFRSF6 (Fas, CD95), TNFRSF25 (DR3), TNFRSF14 (HVEM), TNFSF18, TNFRSF18 (GITR), TNFRSF4 (OX40), TNFSF4 (OX40 Ligand), PD1, PDL1, CTLA4, TNFSF9 (41BB Ligand), TNFRSF9 (41BB), TNFSF14 (LIGHT, HVEM Ligand), TNFSF5 (CD40 Ligand), BTLA, and C
  • an MRD-containing antibody binds to a therapeutic target and a second target that is associated with an escape pathway for resisting the therapeutic effect resulting from targeting the therapeutic target.
  • an MRD-containing antibody binds to EGFR and a target selected from MDR1, cMET, Notch, Notch1, Notch3, Notch4, DLL1, DLL4, Jagged, Jagged1, Jagged2, and Jagged3.
  • the multivalent and monovalent multispecific composition binds 2, 3, 4, 5 or more of these targets.
  • the MRD-containing antibody targets ErbB2 and an angiogenic factor. In specific embodiments, the MRD-containing antibody targets ErbB2 and IGR1R. In another embodiment, the antibody targets ErbB2 and at least one MRD targets an angiogenic factor and/or IGF1R. In one, embodiment, an antibody that binds to the same ErbB2 epitope as trastuzumab is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R. In an additional embodiment, an antibody that competitively inhibits trastuzumab binding is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R.
  • an antibody that comprises the sequences of SEQ ID NOS:59-64 is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R.
  • the trastuzumab antibody is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R.
  • an antibody that binds to ErbB2 is operably linked to an MRD that targets Ang2.
  • the antibody that binds to ErbB2 is linked to an Ang2 binding MRD that binds to, the same Ang2 epitope as an MRD comprising the sequence of MGAQTNFMPMDNDELLLYEQFILQQGLE SEQ ID NO:8.
  • the antibody that binds to ErbB2 is linked to an Ang2 binding MRD that competitively inhibits an MRD comprising the sequence of SEQ ID NO:8.
  • the antibody that binds to ErbB2 is linked to an MRD comprising the sequence of SEQ ID NO:8.
  • At least one Ang2 binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to ErbB2. In some embodiments, at least one Ang2 binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to ErbB2. In some embodiments, at least one Ang2 binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to ErbB2. In some embodiments, at least one Ang2 binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to ErbB2.
  • At least one Ang2 binding MRD is operably linked directly to an antibody that binds to ErbB2. In additional embodiments, at least one Ang2 binding MRD is operably linked to an antibody that binds to ErbB2 via a linker.
  • an antibody that binds to ErbB2 is operably linked to an MRD that targets IGF1R.
  • the antibody that binds to ErbB2 is linked to an IGF1R binding MRD that binds to the same IGF1R epitope as an MRD comprising the sequence of SEQ ID NO:14.
  • the antibody that binds to ErbB2 is linked to an IGF1R binding MRD that competitively inhibits an MRD comprising the sequence of SEQ ID NO:14.
  • the antibody that binds to ErbB2 is linked to an MRD comprising the sequence of SEQ ID NO:14.
  • the antibody that binds ErbB2 is linked to an MRD encoding the sequence SLFVPRPERK (SEQ ID NO:103). In some embodiments, the antibody that binds ErbB2 is linked to an MRD encoding the sequence ESDVLHFTST (SEQ ID NO:104). In some embodiments, the antibody that binds ErbB2 is linked to an MRD encoding the sequence LRKYADGTL (SEQ ID NO:105).
  • At least one IGF1R binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to ErbB2. In some embodiments, at least one IGF1R binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to ErbB2. In some embodiments, at least one IGF1R binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to ErbB2. In some embodiments, at least one IGF1R binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to ErbB2.
  • At least one TGF binding MRD is operably linked directly to an antibody that binds to ErbB2.
  • at least one IGF1R binding MRD is operably linked to an antibody that binds to ErbB2 via a linker.
  • an MRD-containing antibody targets ErbB2 and HER2/3. In some embodiments, an MRD-containing antibody can bind to ErbB2 and HER2/3 simultaneously. In some embodiments, an antibody that binds to ErbB2 is operably linked to an MRD that targets HER2/3. In additional embodiments, at least one HER2/3-binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to ErbB2. In further embodiments, at least one HER2/3-binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to ErbB2.
  • At least one HER2/3-binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to ErbB2. In additional embodiments, at least one HER2/3-binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to ErbB2.
  • At least one HER2/3-binding MRD is operably linked directly to an antibody that binds to ErbB2. In additional embodiments, at least one HER2/3-binding MRD is operably linked to an antibody that binds to ErbB2 via a linker.
  • an MRD-containing antibody targets ErbB2 and HER2/3. In some embodiments, an MRD-containing antibody can bind to ErbB2 and HER2/3 simultaneously. In some embodiments, an antibody that binds to HER2/3 is operably linked to an MRD that targets ErbB2. In additional embodiments, at least one ErbB2-binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to HER2/3. In further embodiments, at least one ErbB2-binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to HER2/3.
  • At least one ErbB2-binding MRD is operably linked to the C-terminus of the light chain of an antibody, that binds to HER2/3. In additional embodiments, at least one ErbB2-binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to HER2/3.
  • At least one ErbB2-binding MRD is operably linked directly to an antibody that binds to HER2/3. In additional embodiments, at least one ErbB2-binding MRD is operably linked to an antibody that binds to HER2/3 via a linker.
  • the MRD-containing antibody targets ErbB2, Ang2, and IGF1R.
  • the MRD-containing antibody comprises an antibody that targets ErbB2, an MRD that targets Ang2, and an MRD that targets IGF1R.
  • the Ang2 and IGF1R MRDs are attached to the same location on the anti-ErbB2 antibody.
  • the Ang2 and IGF1R MRDs are attached to different locations on the anti-ErbB2 antibody.
  • the Ang2 and IGF1R MRDs are on the light chain of the anti-ErbB2 antibody.
  • the Ang2 and IGF1R MRDs are on the heavy chain of the anti-ErbB2 antibody.
  • the Ang2 MRD is on the light chain of the ErbB2 antibody
  • the IGF1R MRD is on the heavy chain of the anti-ErbB2 antibody.
  • the Ang2 MRD is on the heavy chain of the ErbB2 antibody
  • the IGF1R MRD is on the light chain of the anti-ErbB2 antibody.
  • the Ang2 MRD is on the N-terminus of the heavy chain of the ErbB2 antibody
  • the IGF1R MRD is on the C-terminus of the light chain of the anti-ErbB2 antibody.
  • the IGF1R MRD is on the N-terminus of the heavy chain of the ErbB2 antibody
  • the Ang2 MRD is on the C-terminus of the light chain of the anti-ErbB2 antibody.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • multivalent and multispecific compositions comprising an antibody that targets IGF1R, an MRD that targets ErbB2, and an MRD that targets Ang2 are also encompassed by the invention.
  • the MRD-containing antibody targets ErbB2, Ang2, and HER2/3.
  • the MRD-containing antibody comprises an antibody that targets ErbB2, an MRD that targets Ang2, and an MRD that targets HER2/3.
  • the Ang2 and HER2/3 MRDs are attached to the same location on the anti-ErbB2 antibody.
  • the Ang2 and HER2/3 MRDs are attached to different locations on the anti-ErbB2 antibody.
  • the Ang2 and HER2/3 MRDs are on the light chain of the anti-ErbB2 antibody.
  • the Ang2 and HER2/3 MRDs are on the heavy chain of the anti-ErbB2 antibody.
  • the Ang2 MRD is on the light chain of the ErbB2 antibody, and the HER2/3 MRD is on the heavy chain of the anti-ErbB2 antibody. In some embodiments, the Ang2 MRD is on the heavy chain of the ErbB2 antibody, and the HER2/3 MRD is on the light chain of the anti-ErbB2 antibody. In some embodiments, the Ang2 MRD is on the N-terminus of the heavy chain of the ErbB2 antibody, and the HER2/3 MRD is on the C-terminus of the light chain of the anti-ErbB2 antibody.
  • the HER2/3 MRD is on the N-terminus of the heavy chain of the ErbB2 antibody
  • the Ang2 MRD is on the C-terminus of the light chain of the anti-ErbB2 antibody.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • multivalent and multispecific compositions comprising an antibody that targets Ang2, an MRD that targets ErbB2, and an MRD that targets HER2/3
  • multivalent and multispecific compositions comprising an antibody that targets Ang2, an MRD that targets ErbB2, and an MRD that targets HER2/3 are also encompassed by the invention.
  • the MRD-containing antibody targets ErbB2, HER2/3, and IGF1R.
  • the MRD-containing antibody comprises an antibody that targets ErbB2, an MRD that targets HER2/3, and an MRD that targets IGF1R.
  • the HER2/3 and IGF1R MRDs are attached to the same location on the anti-ErbB2 antibody.
  • the HER2/3 and IGF1R MRDs are attached to different locations on the anti-ErbB2 antibody.
  • the HER2/3 and IGF1R MRDs are, on the light chain of the anti-ErbB2 antibody.
  • the HER2/3 and IGF1R MRDs are on the heavy chain of the anti-ErbB2 antibody. In some embodiments, the HER2/3 MRD is on the light chain of the ErbB2 antibody, and the IGF1R MRD is on the heavy chain of the anti-ErbB2 antibody. In some embodiments, the HER2/3 MRD is on the heavy chain of the ErbB2 antibody, and the IGF1R MRD is on the light chain of the anti-ErbB2 antibody. In some embodiments, the HER2/3 MRD is on the N-terminus of the heavy chain of the ErbB2 antibody, and the IGF1R MRD is on the C-terminus of the light chain of the anti-ErbB2 antibody.
  • the IGF1R MRD is on the N-terminus of the heavy chain of the ErbB2 antibody
  • the HER2/3 MRD is on the C-terminus of the light chain of the anti-ErbB2 antibody.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • MRD-containing antibodies comprising an antibody that targets HER2/3, an MRD that targets ErbB2, and an MRD that targets IGF1R
  • multivalent and multispecific compositions e.g., MRD-containing antibodies
  • multivalent and multispecific compositions comprising an antibody that targets IGF1R, an MRD that targets ErbB2, and an MRD that targets HER2/3 are also encompassed by the invention.
  • the MRD-containing antibody targets ErbB2, Ang2, HER2/3, and IGF1R.
  • the MRD-containing antibody comprises an antibody that targets ErbB2, an MRD that targets Ang2, an MRD that targets HER2/3, and an MRD that targets IGF1R.
  • the Ang2, HER2/3, and IGF1R MRDs are attached to the same chain of the anti-ErbB2 antibody.
  • the Ang2, HER2/3, and IGF1R MRDs are attached to different chains of the anti-ErbB2 antibody.
  • the Ang2, HER2/3, and IGF1R MRDs are on the light chain of the anti-ErbB2 antibody.
  • the Ang2, HER2/3, and IGF1R MRDs are on the heavy chain of the anti-ErbB2 antibody. In some embodiments, the Ang2, HER2/3, and IGF1R MRDs are attached to the same terminus of the anti-ErbB2 antibody. In some embodiments, the Ang2, HER2/3, and IGF1R MRDs are attached to different termini of the anti-ErbB2 antibody.
  • Multivalent and multispecific compositions comprising: an antibody that targets HER2/3, an MRD that targets ErbB2, an MRD that targets Ang2, and an MRD that targets IGF1R; multivalent and multispecific compositions (e.g., MRD-containing antibodies) comprising an antibody that targets Ang2, an MRD that targets ErbB2, an MRD that targets HER2/3, and an MRD that targets IGF1R; and multivalent and multispecific compositions (e.g., MRD-containing antibodies) comprising an antibody that targets IGF1R, an MRD that targets ErbB2, an MRD that targets HER2/3, and an MRD that targets Ang2 are also encompassed by the invention.
  • the anti-ErbB2 antibody operably linked to an Ang2 binding MRD binds to both ErbB2 and Ang2 simultaneously. In some embodiments, the anti-ErbB2 antibody operably linked to an IGF1R binding MRD binds to both ErbB2 and IGF1R simultaneously. In some embodiments, the anti-ErbB2 antibody operably linked to a HER2/3 binding MRD binds to both ErbB2 and HER2/3 simultaneously. In some embodiments, the anti-ErbB2 antibody operably linked to an Ang2 MRD, an IGF1R MRD, and/or a HER2/3 MRD binds to ErbB2, Ang2, IGF1R, and/or HER2/3 simultaneously.
  • the anti-ErbB2 antibody operably linked to an Ang2, IGF1R and/or HER2/3 binding MRD(s) exhibits ADCC activity. In additional embodiments, the anti-ErbB2 antibody operably linked to an Ang2, IGF1R, and/or HER2/3 binding MRD(s) down-regulates Akt signaling. In additional embodiments, the anti-ErbB2 antibody operably linked to an Ang2 binding MRD inhibits Ang2 binding to TIE2. In additional embodiments, the anti-ErbB2 antibody operably linked to an IGF1R binding MRD(s) down-regulates IGF1R signaling.
  • the anti-ErbB2 antibody operably linked to an Ang2, IGF1R and/or HER2/3 binding MRD(s) inhibits cell proliferation. In additional embodiments, the anti-ErbB2 antibody operably linked to an Ang2, IGF1R, and/or HER2/3 binding MRD(s) inhibits tumor growth.
  • the MRD-containing antibody targets VEGF and an angiogenic factor. In specific embodiments, the MRD-containing antibody targets VEGF and IGF1R. In another embodiment, the antibody targets VEGF and at least one MRD targets an angiogenic factor and/or IGF1R. In one embodiment, an antibody that binds to the same VEGF epitope as bevacizumab is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R. In an additional embodiment, an antibody that competitively inhibits bevacizumab binding is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R.
  • an antibody that comprises the sequences of SEQ ID NOS:78-79 is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R.
  • the bevacizumab antibody is operably linked to at least one MRD that targets an angiogenic factor and/or IGF1R.
  • an antibody that binds to VEGF is operably linked to an MRD that targets Ang2.
  • the antibody that binds to VEGF is linked to an Ang2 binding MRD that binds to the same Ang2 epitope as an MRD comprising the sequence of SEQ ID NO:8.
  • the antibody that binds to VEGF is linked to an Ang2 binding MRD that competitively inhibits an MRD comprising the sequence of SEQ ID NO:8.
  • the antibody that binds to VEGF is linked to an MRD comprising the sequence of SEQ ID NO:8.
  • At least one Ang2 binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to VEGF. In some embodiments, at least one Ang2 binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to VEGF. In some embodiments, at least one Ang2 binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to VEGF. In some embodiments, at least one Ang2 binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to VEGF.
  • At least one Ang2 binding MRD is operably linked directly to an antibody that binds to VEGF. In additional embodiments, at least one Ang2 binding MRD is operably linked to an antibody that binds to VEGF via a linker.
  • an antibody that binds to VEGF is operably linked to an MRD that targets IGF1R.
  • the antibody that binds to VEGF is linked to an IGF1R binding MRD that binds to the same IGF1R epitope as an MRD comprising the sequence of SEQ ID NO:14.
  • the antibody that binds to VEGF is linked to an IGF1R binding MRD that competitively inhibits an MRD comprising the sequence of SEQ ID NO:14.
  • the antibody that binds to VEGF is linked to an MRD comprising the sequence of SEQ ID NO:14.
  • the antibody that binds ErbB2 is linked to an MRD encoding the sequence SLFVPRPERK (SEQ ID NO:103). In some embodiments, the antibody that binds ErbB2 is linked to an MRD encoding the sequence ESDVLHFTST (SEQ ID NO:104). In some embodiments, the antibody that binds ErbB2 is linked to an MRD encoding the sequence LRKYADGTL (SEQ ID NO:105).
  • At least one IGF1R binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to VEGF. In some embodiments, at least one IGF1R binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to VEGF. In some embodiments, at least one IGF1R binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to VEGF. In some embodiments, at least one IGF1R binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to VEGF.
  • At least one IGF1R binding MRD is operably linked directly to an antibody that binds to VEGF. In additional embodiments, at least one IGF1R binding MRD is operably linked to an antibody that binds to VEGF via a linker.
  • the MRD-containing antibody targets VEGF, Ang2, and IGF1R.
  • the MRD-containing antibody comprises an antibody that targets VEGF, an MRD that targets Ang2, and an MRD that targets IGF1R.
  • the Ang2 and IGF1R MRDs are attached to the same location on the anti-VEGF antibody.
  • the Ang2 and IGF1R MRDs are attached to different locations on the anti-VEGF antibody.
  • the Ang2 and IGF1R MRDs are on the light chain of the anti-VEGF antibody.
  • the Ang2 and IGF1R MRDs are on the heavy chain of the anti-VEGF antibody.
  • the Ang2 MRD is on the light chain of the anti-VEGF antibody, and the IGF1R MRD is on the heavy chain of the anti-VEGF antibody. In some embodiments, the Ang2 MRD is on the heavy chain of the anti-VEGF antibody, and the IGF1R MRD is on the light chain of the anti-VEGF antibody. In some embodiments, the Ang2 MRD is on the N-terminus of, the heavy chain of the anti-VEGF antibody, and the IGF1R MRD is on the C-terminus of the light chain of the anti-VEGF antibody. In some embodiments, the IGF1R MRD is on the N-terminus of the heavy chain of the anti-VEGF antibody, and the Ang2 MRD is on the C-terminus of the light chain of the anti-VEGF antibody.
  • the anti-VEGF antibody operably linked to an Ang2 binding MRD binds to both anti-VEGF and Ang2 simultaneously. In some embodiments, the anti-VEGF antibody operably linked to an IGF1R binding MRD binds to both anti-VEGF and IGFR1 simultaneously. In some embodiments, the anti-VEGF antibody operably linked to an Ang2 binding MRD and an IGF1R binding MRD binds to VEGF, Ang2, and IGF1R simultaneously. In some embodiments, the anti-VEGF antibody operably linked to an Ang2 and/or IGF binding MRD(s) exhibits ADCC activity.
  • the anti-VEGF antibody operably linked to an Ang2 and/or IGF1R binding MRD(s) down-regulates VEGF signaling.
  • the anti-VEGF antibody operably linked to an Ang2 binding MRD inhibits Ang2 binding to TIE2.
  • the anti-VEGF antibody operably linked to an IGF1R binding MRD inhibits IGF1R signaling.
  • the anti-VEGF antibody operably linked to an Ang2 and/or IGF1R binding MRD(s) inhibits cell proliferation.
  • the anti-VEGF antibody operably linked to an Ang2 and/or IGF1R binding MRD(s) inhibits tumor growth.
  • the anti-ErbB2 antibody or the VEGF antibody contains and MRD that inhibits the binding of pertuzumab to ErbB2.
  • an anti-ErbB2 antibody contains at least one MRD that binds to Ang2 or IGF1R and one MRD that inhibits the binding of pertuzumab to ErbB2.
  • an anti-VEGF antibody contains at least one MRD that binds to Ang2 or IGF1R and one MRD that inhibits the binding of pertuzumab to ErbB2.
  • an anti-ErbB2 antibody contains an MRD that binds Ang2, an MRD that binds IGF1R, and an MRD that inhibits the binding of pertuzumab to ErbB2.
  • an anti-VEGF antibody contains an MRD that binds Ang2, an MRD that binds IGF1R, and an MRD that inhibits the binding of pertuzumab to ErbB2.
  • the MRD-containing antibody targets TNF and an angiogenic factor.
  • the antibody targets TNF, and at least one MRD targets an angiogenic factor.
  • an antibody that binds to the same TNF epitope as adalimumab is operably linked to at least one MRD that targets an angiogenic factor.
  • an antibody that competitively inhibits adalimumab binding is operably linked to at least one MRD that targets an angiogenic factor.
  • an antibody that comprises the sequences of SEQ ID NOS:80-85 is operably linked to at least one MRD that targets an angiogenic factor.
  • the adalimumab antibody is operably linked to at least one MRD that targets an angiogenic factor.
  • an antibody that binds to the same TNF epitope as golimumab is operably linked to at least one MRD that targets an angiogenic factor.
  • an antibody that competitively, inhibits golimumab binding is operably linked to at least one MRD that targets an angiogenic factor.
  • the golimumab antibody is operably linked to at least one MRD that targets an angiogenic factor.
  • an antibody that binds to TNF is operably linked to an MRD that targets Ang2.
  • the antibody that binds to TNF is linked to an Ang2 binding MRD that binds to the same Ang2 epitope as an MRD comprising the sequence of SEQ ID NO:8.
  • the antibody that binds to TNF is linked to an Ang2 binding MRD that competitively inhibits an MRD comprising the sequence of SEQ ID NO:8.
  • the antibody that binds to TNF is linked to an MRD comprising the sequence of SEQ ID NO:8.
  • At least one Ang2 binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds to TNF. In some embodiments, at least one Ang2 binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to TNF. In some embodiments, at least one Ang2 binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to TNF. In some embodiments, at least one Ang2 binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to TNF.
  • At least one Ang2 binding MRD is operably linked directly to an antibody that binds to TNF. In additional embodiments, at least one Ang2 binding MRD is operably linked to an antibody that binds to TNF via a linker.
  • the anti-TNF antibody operably linked to an Ang2 binding MRD binds to both TNF and Ang2 simultaneously. In some embodiments, the anti-TNF antibody operably linked to an Ang2 binding MRD exhibits ADCC activity. In additional embodiments, the anti-TNF antibody operably linked to an Ang2 binding MRD inhibits binding of TNF to the p55 and p75 cell surface TNF receptors. In additional embodiments, the anti-TNF antibody operably linked to an Ang2 binding MRD lyses surface TNF-expressing cells in vitro in the presence of complement. In additional embodiments, the anti-TNF antibody operably linked to an Ang2 binding MRD inhibits Ang2 binding to TIE2. In additional embodiments, the anti-TNF antibody operably linked to an Ang2 binding MRD reduces the signs and symptoms of arthritis.
  • the MRD-containing antibody targets TNF and IL6. In some embodiments, the MRD-containing antibody is capable of binding TNF and IL6 simultaneously. Thus, in some embodiments, an antibody that binds to TNF is operably linked to an MRD that targets IL6. In other embodiments, an antibody that binds to IL6 is operably linked to an MRD that targets TNF.
  • At least one IL6-binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds TNF. In some embodiments, at least one IL6-binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to TNF. In some embodiments, at least one IL6-binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to TNF. In some embodiments, at least one IL6-binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to TNF.
  • At least one TNF-binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds IL6. In some embodiments, at least one TNF-binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to IL6. In some embodiments, at least one TNF-binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to IL6. In some embodiments, at least one TNF-binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to IL6.
  • At least one IL6-binding MRD is operably linked directly to an antibody that binds to TNF. In additional embodiments, at least one IL6-binding MRD is operably linked to an antibody that binds to TNF via a linker.
  • At least one TNF-binding MRD is operably linked directly to an antibody that binds to IL6. In additional embodiments, at least one TNF-binding MRD is operably linked to an antibody that binds to IL6 via a linker.
  • the MRD-containing antibody targets TNF and BLyS. In some embodiments, the MRD-containing antibody is capable of binding TNF and BLyS simultaneously. In some embodiments, an antibody that binds to TNF is operably linked to an MRD that targets BLyS. In other embodiments, an antibody that binds to BLyS is operably linked to an MRD that targets TNF.
  • At least one BLyS-binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds TNF. In some embodiments, at least one BLyS-binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to TNF. In some embodiments, at least one BLyS-binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to TNF. In some embodiments, at least one BLyS-binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to TNF.
  • At least one TNF-binding MRD is operably linked to the C-terminus of the heavy chain of an antibody that binds BLyS. In some embodiments, at least one TNF-binding MRD is operably linked to the N-terminus of the heavy chain of an antibody that binds to BLyS. In some embodiments, at least one TNF-binding MRD is operably linked to the C-terminus of the light chain of an antibody that binds to BLyS. In some embodiments, at least one TNF-binding MRD is operably linked to the N-terminus of the light chain of an antibody that binds to BLyS.
  • At least one BLyS-binding MRD is operably linked directly to an antibody that binds to TNF. In additional embodiments, at least one BLyS-binding MRD is operably linked to an antibody that binds to TNF via a linker.
  • At least one TNF-binding MRD is operably linked directly to an antibody that binds to BLyS. In additional embodiments, at least one TNF-binding MRD is operably linked to an antibody that binds to BLyS via a linker.
  • the MRD-containing antibody targets Ang2, TNF, and IL6. In some embodiments, the MRD-containing antibody is capable of binding Ang2, TNF, and IL6 simultaneously. In some embodiments, an antibody that binds to TNF is operably linked to an MRD that targets Ang2 and an MRD that targets IL6. In some embodiments, the Ang2 and IL6-binding MRDs are located on the same antibody chain. In some embodiments, the Ang2 and IL6-binding MRDs are located on the same antibody terminus. In some embodiments, the Ang2 and IL6-binding MRDs are located on different antibody chains. In some embodiments, the Ang2 and IL6-binding MRDs are located on different antibody termini.
  • an antibody that binds to Ang2 is operably linked to an MRD that targets TNF and an MRD that targets IL6.
  • the TNF and IL6-binding MRDs are located on the same antibody chain. In some embodiments, the TNF and IL6-binding MRDs are located on the same antibody terminus. In some embodiments, the TNF and IL6-binding MRDs are located on different antibody chains. In some embodiments, the TNF and IL6-binding MRDs are located on different antibody termini.
  • an antibody that binds to IL6 is operably linked to an MRD that targets Ang2 and an MRD that targets TNF.
  • the Ang2 and TNF-binding MRDs are located on the same antibody chain.
  • the Ang2 and TNF-binding MRDs are located on the same antibody terminus.
  • the Ang2 and TNF-binding MRDs are located on different antibody chains.
  • the Ang2 and TNF-binding MRDs are located on different antibody termini.
  • the MRD-containing antibody targets Ang2, TNF, and BLyS. In some embodiments, the MRD-containing antibody is capable of binding Ang2, TNF, and BLyS simultaneously. In some embodiments, an antibody that binds to TNF is operably linked to an MRD that targets Ang2 and an MRD that targets BLyS. In other embodiments, an antibody that binds to BLyS is operably linked to an MRD that targets TNF and an MRD that targets Ang2. In other embodiments, an antibody that binds to Ang2 is operably linked to an MRD that targets TNF and an MRD that targets BLyS.
  • the Ang2, BLyS, and/or TNF-binding MRDs are located on the same antibody chain. In some embodiments, Ang2, BLyS, and/or TNF-binding MRDs are located on the same antibody terminus. In some embodiments, the Ang2, BLyS, and/or TNF-binding MRDs are located on different antibody chains. In some embodiments, the Ang2, BLyS, and/or TNF-binding MRDs are located on different antibody termini.
  • the MRD-containing antibody targets Ang2, TNF, IL6, and BLyS. In some embodiments, the MRD-containing antibody is capable of binding Ang2, TNF, IL6 and BLyS simultaneously. In some embodiments, an antibody that binds to TNF is operably linked to an MRD that targets Ang2, an MRD that, targets IL6, and an MRD that targets BLyS. In some embodiments, an antibody that binds to Ang2 is operably linked to an MRD that targets TNF, an MRD that targets IL6, and an MRD that targets BLyS.
  • an antibody that binds to IL6 is operably linked to an MRD that targets Ang2, an MRD that targets TNF, and an MRD that targets BLyS.
  • an antibody that binds to BLyS is operably linked to an MRD that targets Ang2, an MRD that targets IL6, and an MRD that targets TNF.
  • the TNF, Ang2, IL6, and/or BLyS-binding MRDs are located on the same antibody chain.
  • the TNF, Ang2, IL6 and/or BLyS-binding MRDs are located on the same antibody terminus.
  • the TNF, Ang2, IL6, and/or BLyS-binding MRDs are located on different antibody chains. In some embodiments, the TNF, Ang2, IL6 and/or BLyS-binding MRDs are located on different antibody termini.
  • the multivalent and multispecific compositions of the invention can be produced by any method known in the art for the synthesis of antibodies, polypeptides, immunoconjugates, and cytotoxins, in particular, by chemical synthesis or by recombinant expression techniques.
  • An advantage of multivalent and multispecific compositions is that they can be produced using protocols that are known in the art for producing antibodies.
  • the antibody-MRD fusion molecules can be encoded by a polynucleotide comprising a nucleotide sequence.
  • the polynucleotides described herein can encode an MRD, an antibody heavy chain, an antibody light chain, a fusion protein comprising an antibody heavy chain and at least one MRD, and/or a fusion protein comprising an antibody light chain and at least one MRD.
  • the invention provides vector constructs comprising a polynucleotide sequence(s) encoding multivalent and multispecific compositions (e.g., MRD-containing antibodies) and a host cell comprising these rector constructs.
  • a polynucleotide sequence(s) encoding multivalent and multispecific compositions e.g., MRD-containing antibodies
  • Standard techniques for cloning and transformation may be used in the preparation of cell lines expressing the multivalent and multispecific compositions (e.g. MRD-containing antibodies) of the invention.
  • Recombinant expression vectors containing a polynucleotide sequence(s) encoding multivalent and multispecific compositions (e.g., MRD-containing antibodies) of the invention can be prepared using well known techniques.
  • the expression vectors include a polynucleotide coding sequence operably linked to suitable transcriptional or translational regulatory nucleotide sequences such as, those derived from mammalian, microbial, viral, or insect genes.
  • suitable transcriptional or translational regulatory nucleotide sequences such as, those derived from mammalian, microbial, viral, or insect genes.
  • Exemplary regulatory sequences present in the expression vector constructs include transcriptional promoters, operators, enhancers, mRNA ribosomal binding sites, and/or other appropriate sequences which control transcription and translation initiation and termination.
  • Nucleotide sequences are “operably linked” when the regulatory sequence functionally relates to the nucleotide sequence for the appropriate polypeptide.
  • a promoter sequence is operably linked to, for example, an antibody heavy chain-MRD sequence if the promoter nucleotide sequence controls the transcription of the appropriate nucleotide sequence.
  • the polynucleotide coding sequence in the expression vector can include additional heterologous sequences encoding polypeptides such as, signal peptides that are not naturally associated with antibody heavy and/or light chain sequences.
  • a nucleotide sequence for a signal peptide secretory leader
  • a signal peptide that is functional in the intended host cells enhances extracellular secretion of the appropriate antibody.
  • the signal peptide can be cleaved from the polypeptide upon secretion of antibody from the cell. Examples of sequences encoding secretory signals that can be included in the expression vectors include those described in for example, U.S. Pat. Nos. 5,698,435, 5,698,417, and 6,204,023.
  • a variety of host-expression vector systems can be utilized to express the coding sequence an MRD-containing antibody.
  • Host cells useful in the present invention include but are not limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis ) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia ) transformed with recombinant yeast expression vectors containing, antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., Baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing MRD-containing antibody coding sequences.
  • microorganisms such as bacteria (e.g., E. coli, B. subtilis ) transformed with re
  • the mammalian cell systems are used to produce the multivalent and multispecific compositions of the invention (e.g., MRD-containing antibodies).
  • Mammalian cell systems typically utilize recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • mammalian host cells useful for producing the multivalent and multispecific compositions of the invention include, CHO cells, BHK cells, NS0 cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells, COS cells, 293 cells, 3T3 cells and hybridoma cells.
  • Vectors containing the polynucleotides encoding the multivalent and multispecific compositions of the invention include plasmid vectors, a single and double-stranded phage vectors, as well as single and double-stranded RNA or DNA viral vectors.
  • the vectors can be routinely introduced into host cells using known techniques for introducing DNA and RNA into cells.
  • Phage and viral, vectors may also be introduced into host cells in the form of packaged or encapsulated virus using known techniques for infection and transduction.
  • viral vectors may be replication competent or alternatively, replication defective.
  • cell-free translation systems may also be used to produce the protein using RNAs derived from the DNA expression constructs of the invention (see, e.g., Intl. Appl. Publ. WO86/05807 and WO89/01036; and U.S. Pat. No. 5,122,464).
  • an MRD-containing antibody comprising: culturing a host cell comprising one or more polynucleotides or an expression vector comprising one or more isolated polynucleotides in a medium under conditions allowing the expression of said one or more polynucleotide, wherein said one or more polynucleotides encodes one or more polypeptides that form part of MRD-containing antibody; and recovering said MRD-containing antibody.
  • Prokaryotes useful as host cells in producing the compositions of the invention include gram negative or gram positive organisms such as, E. coli and B. subtilis .
  • Expression vectors for use in prokaryotic host cells generally contain one or more phenotypic selectable marker genes (e.g., genes encoding proteins that confer antibiotic resistance or that supply an autotrophic requirement).
  • prokaryotic host expression vectors examples include the pKK223-3 (Pharmacia, Uppsala, Sweden), pGEM1 (Promega, Wis., USA), pET (Novagen, Wis., USA) and pRSET (Invitrogen, Calif., USA) series of vectors (see, e.g., Studier, J. Mol. Biol, 219:37 (1991) and Schoepfer, Gene 124:83 (1993)).
  • Exemplary promoter sequences frequently used in prokaryotic host cell expression vectors include T7, (Rosenberg et al., Gene 56: 125-135 (1987)), beta-lactamase (penicillinase), lactose promoter system (Chang et al., Nature 275:615 (1978)); and Goeddel et al., Nature 281:544 (1979)), tryptophan (tip) promoter system (Goeddel et al., Nucl. Acids Res. 8:4057, (1980)), and tac promoter (Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.).
  • eukaryotic host cell systems can be used, including yeast cells transformed with recombinant yeast expression vectors containing the coding sequence of an MRD-containing antibody of the present invention, such as, the expression systems taught in U.S. Pat. Appl. No. 60/344,169 and WO03/056914 (methods for producing human-like glycoprotein in a non-human eukaryotic host cell) (the contents of each of which are incorporated by reference in their entirety).
  • Exemplary yeast that can be used to produce compositions of the invention, such as, MRDs include yeast from the genus Saccharotnyces, Pichia, Actinomycetes and Kluyveromyces .
  • Yeast vectors typically contain an origin of replication sequence from a 2 mu yeast plasmid, an autonomously replicating sequence (ARS), a promoter region, sequences for polyadenylation, sequences for transcription termination, and a selectable marker gene.
  • ARS autonomously replicating sequence
  • promoter sequences in yeast expression constructs include promoters from metallothionein, 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem.
  • glycolytic enzymes such as, enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • Additional suitable vectors and promoters for use in yeast expression as well as yeast transformation protocols are known in the art. See, e.g., Fleer et al., Gene, 107:285-195 (1991) and Hinnen et al., Proc. Natl. Acad. Sci., 75:1929 (1978).
  • Insect and plant host cell culture systems are also useful for producing the compositions of the invention.
  • host cell systems include for example, insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the coding sequence of an MRD-containing antibody; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing the coding sequence of an MRD-containing antibody, including, but not limited to, the expression systems taught in U.S. Pat. No. 6,815,184, WO2004/057002, WO2004/024927, U.S. Pat. Appl. Nos. 60/365,769, 60/368,047, and WO2003/078614, the contents of each of which is herein incorporated by reference in its entirety.
  • eukaryotic host cell systems including animal cell systems infected with recombinant virus expression vectors (e.g., adenovirus, vaccinia virus) including cell lines engineered to contain multiple copies of the DNA encoding an MRD-containing antibody either stably amplified (CHO/dhfr) or unstably amplified in double-minute chromosomes (e.g., murine cell lines).
  • virus expression vectors e.g., adenovirus, vaccinia virus
  • the vector comprising the polynucleotide(s) encoding the MRD-containing antibody of the invention is polycistronic.
  • Exemplary mammalian cells useful for producing these compositions include 293 cells (e.g., 293T and 293F), CHO cells, BHK cells, NS0 cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 (Crucell, Netherlands) cells or hybridoma cells, other mammalian cells.
  • Additional exemplary mammalian host cells that are useful in practicing the invention include but are not limited, to VERY, Hela, COS, MDCK, 3T3, W138, BT483, Hs578T, HTB2, BT20 and T47D, CRL7O3O and HsS78Bst cells.
  • Transcriptional and translational control sequences for mammalian host cell expression vectors are frequently derived from viral genomes.
  • Commonly used promoter sequences and enhancer sequences in mammalian expression vectors include, sequences derived from Polyoma virus, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus (CMV).
  • Exemplary commercially available expression vectors for use in mammalian host cells include pCEP4 (Invitrogen®) and pcDNA3 (Invitrogen®).
  • a number of selection systems can be used in, mammalian host-vector expression systems, including, but not limited to, the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes, which can be employed in tk, hgprt ⁇ or aprt ⁇ cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for e.g., dhfr, gpt, neo, hygro, trpB, hisD, ODC (ornithine decarboxylase), and the glutamine synthase system.
  • a variety of host-expression vector systems may be utilized to express the coding sequence an MRD-containing antibody.
  • a host cell strain can be chosen which modulates the expression of inserted antibody sequences, or modifies and processes the antibody gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products can be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the antibody or portion thereof expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Stable expression typically achieves more reproducible results than transient expression and also is more amenable to large-scale production; however, it is within the skill of one in the art to determine whether transient expression is better for a particular situation.
  • host cells can be transformed with the respective coding nucleic acids controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows selection of cells which have stably integrated the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • the multivalent and multispecific compositions are expressed at levels (titers) comparable to those of antibodies.
  • the multivalent and multispecific compositions are expressed at least about 10 ⁇ g/ml at least about 20 ⁇ g/ml, or at least about 30 ⁇ g/ml.
  • the multivalent and multispecific compositions are expressed at least about 40 ⁇ g/ml or at least about 50 ⁇ g/ml.
  • the multivalent and multispecific compositions are expressed at least about 60 ⁇ g/ml, at least about 70 ⁇ g/ml, at least about 80 ⁇ g/ml, at least about 90 ⁇ g/ml, at least about 95 ⁇ g/ml, at least about 100 ⁇ g/ml, at least about 110 ⁇ g/ml, at least about 120 ⁇ g/ml, at least about 130 ⁇ g/ml, at least about 140 ⁇ g/ml, at, least about 150 ⁇ g/ml, at least about 160 ⁇ g/ml, at least about 170 ⁇ g/ml, at least about 180 ⁇ g/ml, at least about 190 ⁇ g/ml, or at least about 200 ⁇ g/ml.
  • the expression levels of an antibody molecule can be increased by vector amplification and the use recombinant methods and tools known in the art, including chromatin remodeling strategies to, enhance transgene, expression.
  • the present invention is further directed to a method for modifying the glycosylation profile of an MRD-containing antibody that is produced by a host cell, comprising expressing in said host cell a nucleic acid encoding an MRD-containing antibody and a nucleic acid encoding a polypeptide with a glycosyltransferase activity, or a vector comprising such nucleic acids.
  • Genes with glycosyltransferase activity include ⁇ (1,4)-N-acetylglucosaminyltransferase III (GnTII), ⁇ -mannosidase II (ManII), ⁇ (1,4)-galactosyltransferase (GalT), ⁇ (1,2)-N-acetylglucos aminyltransferase I (GnTI), and ⁇ (1,2)-N-acetylglucosaminyltransferase II (GnTII).
  • a combination of genes with glycosyltransferase activity are expressed in the host cell (e.g., GnTIII and Man II).
  • the method also encompasses expression of one or more polynucleotide(s) encoding the MRD-containing antibody in a host cell in which a glycosyltransferase gene has been disrupted or otherwise deactivated (e.g., a host cell in which the activity of the gene encoding ⁇ 1-6 core fucosyltransferase has been knocked out).
  • the MRD-containing antibody can be produced in a host cell that further expresses a polynucleotide encoding a polypeptide having GnTIII activity to modify the glycosylation pattern.
  • the polypeptide having GnTIII activity is a fusion polypeptide comprising the Golgi localization domain of a Golgi resident polypeptide.
  • the expression of the MRD-containing antibody in a host cell that expresses a polynucleotide encoding a polypeptide having GnTIII activity results in an MRD-containing antibody with increased Fc receptor binding affinity and increased effector function.
  • the present invention is directed to a host cell comprising (a) an isolated nucleic acid comprising a sequence encoding a polypeptide having GnTIII activity; and (b) an isolated polynucleotide encoding an MRD-containing antibody of the present invention, such as, a chimeric, primatized or humanized antibody.
  • the polypeptide having GnTIII activity is a fusion polypeptide comprising the catalytic domain of GnTIII and the Golgi localization domain is the localization domain of mannosidase II.
  • the multivalent and multispecific compositions with altered glycosylation produced by the host cells of the invention typically exhibit increased Fc receptor binding, affinity and/or increased effector function as a result of the modification of the host cell (e.g., by expression of a glycosyltransferase gene).
  • the increased Fc receptor binding affinity can be increased binding to an Fey activating receptor, such as, the Fc ⁇ RIIIa receptor.
  • the increased effector function can be an increase in one or more of the following: increased antibody-dependent cellular cytotoxicity, increased antibody-dependent cellular phagocytosis (ADCP), increased cytokine secretion, increased immune-complex-mediated antigen uptake by antigen-presenting cells, increased Fc-mediated cellular cytotoxicity, increased binding to NK cells, increased binding to macrophages, increased binding to polymorphonuclear cells (PMNs), increased binding to monocytes, increased crosslinking of target-bound antibodies, increased direct signaling inducing apoptosis, increased dendritic cell maturation, and increased T cell, priming.
  • ADCP antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • cytokine secretion increased immune-complex-mediated antigen uptake by antigen-presenting cells
  • Fc-mediated cellular cytotoxicity increased binding to NK cells
  • macrophages increased binding to macrophages
  • PMNs polymorphonuclear cells
  • a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • it can be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • the multivalent and multispecific compositions of the present invention or fragments thereof are optionally fused to heterologous polypeptide sequences described herein or otherwise known in the art to facilitate purification.
  • the multivalent and multispecific compositions or fragments thereof are optionally fused to heterologous polypeptide sequences described herein or otherwise known in the art to facilitate purification. More particularly, it is envisioned that ligands (e.g., antibodies and other affinity matrices) for MRDs or other components of the multivalent and multispecific compositions can be used in affinity columns for affinity purification and that optionally, the MRDs or other components of the multivalent and monovalent multispecific composition that are bound by these ligands are removed from the composition prior to final preparation of the multivalent and multispecific compositions using techniques known in the art.
  • ligands e.g., antibodies and other affinity matrices
  • the multivalent and multispecific compositions (e.g., MRD-containing antibodies) described herein are useful in a variety of applications including, but not limited to, therapeutic treatment methods, such as, the treatment of cancer.
  • the multivalent and multispecific compositions (e.g., MRD-containing antibodies) are useful for inhibiting tumor growth, reducing neovascularization, reducing angiogenesis, inducing differentiation, reducing tumor volume, and/or reducing the tumorigenicity of a tumor.
  • the methods of use may be in vitro, ex vivo, or in vivo methods. Cancer therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the
  • the PDR discloses dosages of the agents that have been used in treatment of various cancers.
  • the dosing regimen and dosages of these aforementioned chemotherapeutic drugs that are therapeutically effective will depend on the particular cancer being treated, the extent of the disease and other factors familiar to the physician of skill in the art and can be determined by the physician.
  • the contents of the PDR are expressly incorporated herein in its entirety by reference.
  • the 2006 edition of the Physician's Desk Reference (PDR) discloses the mechanism of action and preferred doses of treatment and dosing schedules for thalidomide (p 979-983), VELCADE® (p 2102-2106) and melphalan (p 976-979).
  • the multivalent and multispecific compositions are formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the dosage ranges for the administration of the multivalent and multispecific compositions of the invention are those large enough to produce the desired effect in which the disease symptoms mediated by the target molecule are ameliorated.
  • the dosage should not be so large as to cause adverse side effects, such as, hyperviscosity syndromes, pulmonary edema, congestive heart failure, and the like.
  • the dosage will vary with the age, condition, sex and extent of the disease in the patient and can be determined by one of skill in the art.
  • the dosage can be adjusted by the individual physician in the event of any complication.
  • compositions that contains active ingredients dissolved or dispersed therein are well understood in the art.
  • compositions are prepared as sterile injectables either as liquid solutions or suspensions, aqueous or nonaqueous.
  • solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared.
  • the preparation can also be emulsified.
  • an antibody-MRD containing composition can take the form of solutions, suspensions, tablets, capsules, sustained release formulations or powders, or other compositional forms.
  • the compositions of the invention are formulated to ensure or optimize distribution in vivo.
  • the blood-brain barrier excludes many highly hydrophilic compounds and if so desired, the compositions are prepared so as to increase transfer across the BBB, by for example, formulation in liposomes.
  • the liposomes may comprise one or more moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., Ranade Clin. Pharmacol. 29:685 (1989)).
  • the active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof.
  • Physiologically tolerable carriers are well known in the art.
  • composition of the present invention can include pharmaceutically acceptable salts of the components therein.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like
  • Liquid compositions can also contain liquid phases in addition to and to the exclusion of water.
  • additional liquid phases are glycerin, vegetable oils such as, cottonseed oil, organic esters such as, ethyl oleate, and water-oil emulsions.
  • a therapeutic composition contains a multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) of the present invention, typically in an amount of at least 0.1 weight percent of MRD-containing antibody fusion per weight of total therapeutic composition.
  • a weight percent is a ratio by weight of MRD-containing antibody per total composition.
  • 0.1 weight percent is 0.1 grams of MRD-containing antibody per 100 grams of total composition.
  • the MRD-containing antibody are formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the dosage schedule and amounts effective for therapeutic and prophylactic uses i.e., the “dosing regimen” will depend upon a variety of factors, including the cause, stage and severity of the disease or disorder, the health, physical status, age of the mammal being treated, and the site and mode of the delivery of the MRD-containing antibody.
  • Therapeutic efficacy and toxicity of the complex and formation can be determined by standard pharmaceutical, pharmacological, and toxicological procedures in cell cultures or experimental animals. Data obtained from these procedures can likewise be used in formulating a range of dosages for human use.
  • therapeutic index i.e., the dose therapeutically effective in 50 percent of the population divided by the dose lethal to 50 percent of the population (ED 50 /LD 50 )
  • the dosage is preferably within a range of concentrations that includes the ED 50 with little or no toxicity, and may vary within this range depending on the dosage form employed, sensitivity of the patient, and the route of administration.
  • the dosage regimen also takes into consideration pharmacokinetics parameters known in the art, such as, drug absorption rate, bioavailability, metabolism and clearance (see, e.g., Hidalgo-Aragones, J. Steloid Biochem. Mol. Biol. 58:611-617 (1996); Groning et al., Pharmazie 51:337-341 (1996); Fotherby Contraception 54:59-69 (1996); and Johnson et al., J. Pharm. Sci. 84:1144-1146 (1995)). It is well within the state of the art for the clinician to determine the dosage regimen for each subject being treated.
  • a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • a multivalent and monovalent multispecific composition can be administered depending on the dosage and frequency as required and tolerated by the subject.
  • the duration of prophylactic and therapeutic treatment will vary depending on the particular disease or condition being treated. Some diseases are amenable to acute treatment whereas others require long-term, chronic therapy.
  • MRD-containing antibody can be administered serially, or simultaneously with the additional therapeutic agent.
  • Therapeutically effective amounts of MRD-containing antibody of the invention vary according to, for example, the targets of the MRD-containing antibody and the potency of conjugated cytotoxic agents encompassed by various embodiments of the invention
  • therapeutically effective dose of an a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • a soluble ligand such as, TNF alpha
  • a multivalent and multispecific compositions comprising a maytansinoid cytotoxic agent are likely to be lower than the dosage of an a multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) comprising a less potent chemotherapeutic, such as, taxol, or the counterpart a multivalent and monovalent multispecific composition does not contain a cytotoxic agent.
  • a therapeutically effective dose of an a multivalent and monovalent multispecific composition is an amount selected from about 0.00001 mg/kg to about 20 mg/kg, from about 0.00001 mg/kg to about 10 mg/kg, from about 0.00001 mg/kg to about 5 mg/kg, from about 0.0001 mg/kg to about 20 mg/kg, from about 0.0001 mg/kg to about 10 mg/kg, from about 0.0001 mg/kg to about 5 mg/kg, from about 0.001 mg/kg to about 20 mg/kg, from about 0.001 mg/kg to about 10 mg/kg, and from about 0.001 mg/kg to about 5 mg/kg of the patient's body weight, in one or more dose administrations daily, for one or several days.
  • an a multivalent and monovalent multispecific composition e.g., MRD-containing antibody
  • a therapeutically effective amount of an a multivalent and monovalent multispecific composition is an amount such that when administered in a physiologically tolerable composition is sufficient to achieve a plasma concentration of from about 0.1 microgram ( ⁇ g) per milliliter (ml) to about 100 ⁇ g/ml, from about 1 ⁇ g/ml to about 5 ⁇ g/ml, and usually about 5 ⁇ g/ml.
  • the dosage can vary from about 0.1 mg/kg to about 300 mg/kg, from about 0.2 mg/kg to about 200 mg/kg, from about 0.5 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or several days.
  • the a multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) is administered at about 1 mg/kg to about 50 mg/kg, about 1 mg/kg to about 25 mg/kg, about 1 mg/kg to about 20 mg/kg, about 1 mg/kg to about 15 mg/kg, about 1 mg/kg to about 10 mg/kg, or about 1 mg/kg to about 5 mg/kg.
  • the interval between dose administration of the multivalent and monovalent multispecific composition is about daily, about twice a week, about every week, about every other week, or about every three weeks.
  • the multivalent and monovalent multispecific composition is administered first at a higher loading dose and subsequently at a lower maintenance dose.
  • therapeutic composition comprise multivalent and multispecific compositions (e.g., MRD-containing antibodies) in an amount of at least 0.1 weight percent of antibody per weight of total therapeutic composition.
  • a weight percent is a ratio by weight of antibody/total composition.
  • 0.1 weight percent is 0.1 grams of antibody-MRD per 100 grams of total composition.
  • a therapeutic composition comprising a multivalent and monovalent multispecific composition contains about 10 micrograms ( ⁇ g) per milliliter (ml) to about 100 milligrams (mg) per ml of antibody as active ingredient per volume of composition.
  • a therapeutic composition comprising a multivalent and monovalent multispecific composition contains about 1 mg/ml to about 10 mg/ml (i.e., about 0.1 to 1 weight percent) of antibody as active ingredient per volume of composition.
  • a multivalent and multispecific composition e.g., an MRD containing antibody
  • an antibody-MRD is administered in a dosing concentration and regimen that is the same as the antibody component of the antibody-MRD molecule alone (e.g., a commercial antibody, or a so-called “biosimilar” or a “biobetter” thereof).
  • the multivalent and multispecific composition can have a different PK profile from a corresponding antibody.
  • the dosing concentration is expected to be less than that of the corresponding antibody.
  • therapeutically effective dosing concentrations and regimens for these compositions can routinely be determined using factors and criteria known in the art.
  • the multivalent and multispecific compositions need not be, but optionally are, formulated with one or more agents currently used to prevent or treat the disorder in question.
  • the effective amount of such other agents depends on the amount of multivalent and monovalent multispecific composition present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • the appropriate dosage of the multivalent and monovalent multispecific composition (e.g., MRD-containing antibody) will depend on the type of disease to be treated, as defined above, the severity and course of the disease, previous therapy, the patient's clinical history, and the discretion of the attending physician.
  • the multivalent and monovalent multispecific composition is suitably administered to the patient at one time or over a series of treatments.
  • the multivalent and monovalent multispecific composition is administered by intravenous infusion or by subcutaneous injections.
  • the multivalent and monovalent multispecific composition is administered parenterally by injection or by gradual infusion over time.
  • the target molecule can typically be accessed in the body by systemic administration and therefore most often treated by intravenous administration of therapeutic compositions, other tissues and delivery means are contemplated where there is likelihood that the tissue targeted contains the target molecule.
  • the multivalent and monovalent multispecific composition can be administered intravenously, intraperitoneally, intramuscularly, subcutaneously, intracavity, transdermally, and can be delivered by peristaltic means. Multivalent and multispecific compositions can also be delivered by aerosol to airways and lungs.
  • the antibody-MRD molecule is administered by intravenous infusion.
  • the antibody-MRD molecule is administered by subcutaneous injection.
  • the therapeutic compositions containing a multivalent and monovalent multispecific composition can conventionally be administered intravenously, as by injection of a unit dose, for example.
  • unit dose when used in reference to a therapeutic composition of the present invention refers to physically discrete units suitable as unitary dosage for the patient, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent; i.e., carrier, or vehicle.
  • the therapeutic compositions containing a human monoclonal antibody or a polypeptide are administered subcutaneously.
  • compositions of the invention are administered in a manner compatible with the dosage formulation, and in a therapeutically effective amount.
  • quantity to be administered depends on the patient to be treated, capacity of the patient's system to utilize the active ingredient, and degree of therapeutic effect desired. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner and are peculiar to each individual.
  • suitable dosage ranges for systemic application are disclosed herein and depend on the route of administration. Suitable regimes for administration are also variable but are typified by an initial administration followed by repeated doses at one or more hour intervals by a subsequent injection or other administration. Alternatively, continuous intravenous infusion sufficient to maintain concentrations in the blood in the ranges specified for in vivo therapies are contemplated.
  • the invention provides a method for treating or preventing a disease, disorder, or injury comprising administering a therapeutically effective amount or prophylactically effective amount of antibody-MRD molecule to a patient in need thereof.
  • the disease, disorder or injury is cancer.
  • the disease, disorder or injury is a disease or disorder of the immune system, such as, inflammation or an autoimmune disease.
  • Multivalent and multispecific compositions are expected to have at least the same therapeutic efficacy as the antibody contained in the MRD antibody containing antibody when administered alone. Accordingly, it is envisioned that the multivalent and multispecific compositions (e.g., MRD-containing antibodies) can be administered to a patient to treat or prevent a disease, disorder, or injury for which the antibody contained in the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD-containing antibody, demonstrates a reasonably correlated beneficial activity in treating or preventing such disease, disorder or injury. This beneficial activity can be demonstrated in vitro, in an in vivo animal model, or in human clinical trials.
  • an MRD-containing antibody is administered to a patient to treat or prevent a disease, disorder or injury for which the antibody component of the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD-containing antibody, demonstrates therapeutic or prophylactic efficacy in vitro or in an animal model.
  • an MRD-containing antibody is administered to a patient to treat or prevent a disease, disorder or injury for which the antibody component of the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD-containing antibody, demonstrates therapeutic or prophylactic efficacy in humans.
  • an MRD-containing antibody is administered to a patient to treat or prevent a disease, disorder or injury for which the antibody component of the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD-containing antibody, has been approved by a regulatory authority for use in such treatment or prevention.
  • an MRD-containing antibody is administered in combination with another therapeutic to treat or prevent a disease, disorder or injury for which the antibody component of the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD antibody, in combination with the therapeutic, or a different therapeutic that functions in the same way as the therapeutic in the combination, demonstrates therapeutic or prophylactic efficacy in vitro or in an animal model.
  • an MRD-containing antibody is administered in combination with another therapeutic to treat or prevent a disease, disorder or injury for which the antibody component of the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD antibody, in combination with the therapeutic, or a different therapeutic that functions in the same way as the therapeutic in the combination, demonstrates therapeutic or prophylactic efficacy in humans.
  • an MRD-containing antibody is administered in combination with another therapeutic to treat or prevent a disease, disorder or injury for which the antibody component of the MRD-containing antibody, or an antibody that functions in the same way as the antibody contained in the MRD antibody, in combination with the therapeutic, or a different therapeutic that functions in the same way as the therapeutic in the combination, has been approved by a regulatory authority for use in such treatment or prevention.
  • the administration of an MRD-containing antibody in combination with more than one therapeutic as described above is also encompassed by the invention.
  • an MRD-containing antibody is administered in combination with a compound that promotes apoptosis, inhibits apoptosis, promotes cell survival, inhibits cell survival, promotes senescence of diseased or aberrant cells, inhibits cell senescence, promotes cell proliferation, inhibits cell proliferation, promotes cell differentiation, inhibits cell differentiation, promotes cell activation, inhibits cell activation, promotes cell metabolism, inhibits cell metabolism, promotes cell adhesion, inhibits cell adhesion, promotes cell cycling or cell division, inhibits cell cycling or cell division, promotes DNA replication or repair, inhibits DNA replication or repair, promotes transcription or translation, or inhibits transcription or translation.
  • an MRD-containing antibody is administered in combination with a compound that promotes apoptosis or senescence of diseased or aberrant cells.
  • the MRD-containing antibody is administered in combination with a compound that agonizes, antagonizes or reduces the activity of: EGFR, ErbB2, cMET, TNFa, TGFb, integrin ⁇ v ⁇ 3, TLR2, TLR3, TLR4, TLR5, TLR7, TLR8, TLR9, TNFR1, TNFRSF10A (TRAIL R1 DR4), TNFRSF10B (TRAIL R2 DR5), TNF, TRAIL, IFN beta, MYC, Ras, BCR, ABL, JNK, CKH2, CHK1, CDK1, RAC1, MEK, MOS, mTOR, AKT, NFkB, Ikk, IAP1, IAP2, XIAP, b-catenin, survivin, HDAC, HSP70, HSP90
  • an MRD-containing antibody is administered in combination with a compound that inhibits cell survival.
  • the MRD-containing antibody is administered in combination with a compound that antagonizes or reduces the activity of: VEGF, VEGFR1, VEGFR2, IGF1R, IGF1, IGF2, PDGF-A, PDGF-B, PDGF-CC, PDGF-C, PDGF-D, PDGFRA, PDGFRB, TFGa, TGFB3, PI3K, TNFSF13B (BLYS), TNFRSF13C (BAFFR), INK, NFKB, SIP, integrin ⁇ v ⁇ 3, or survivin.
  • an MRD-containing antibody is administered in combination with a compound that regulates cell proliferation.
  • the MRD-containing antibody is administered in combination with a compound that antagonizes or reduces the activity of: VEGF, VEGFR, EGFR, ErbB2, NFKB, HIF, MUC1, MUC2, or HDAC.
  • an MRD-containing antibody is administered in combination with a compound that regulates cell adhesion.
  • the MRD-containing antibody is administered in combination with a compound that inhibits or reduces the activity of: MMP1, MMP2, MMP7, MMP9, MMP12, PLAU, ⁇ v ⁇ 1 integrin, ⁇ v ⁇ 3 integrin, ⁇ v ⁇ 5 integrin, TGFb, EPCAM, ⁇ 1 ⁇ 1 integrin, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 2 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ 9 ⁇ 1 integrin, ⁇ 6 ⁇ 4 integrin, ⁇ M ⁇ 2 integrin, CEA, L1, Mel-CAM, or HIF1.
  • the MRD-containing antibody is administered in combination with a compound that inhibits or reduces the activity of ⁇ v ⁇ 3 integrin, ⁇ v ⁇ 5 integrin, or ⁇ 5 ⁇ 1 integrin.
  • the MRD-containing antibody is administered in combination with: MEDI-522 (VITAXIN, Abegrin; MedImmune), ATN-161 (Attenuon), EMD 121974 (Merck KGaA), CNTO 95 (Cenotocor), or velociximab (M200, Protein Design Labs).
  • an MRD-containing antibody is administered in combination with a compound that regulates cell activation.
  • the MRD-containing antibody is administered in combination with a compound that promotes, inhibits or reduces the activity of: CD80, CD86, MHC, PDL2 (B7-DC), B7-H1, B7-H2 (ICOSL), B7-H3, B7-H4, CD28, CTLA4, TCR, PD1, CD80, or ICOS.
  • an MRD-containing antibody is administered in combination with a compound that regulates cell cycling, cell division or mitosis.
  • the MRD-containing antibody is administered in combination with a compound that antagonizes or reduces the activity of PI3K, SMO, Ptch, HH, SHH, plk1, plk2, plk3, plk4, aurora A, aurora B, aurora C, CDK1, CDK2, CDK4, CHK1, CHK2, GSK3B, PAK, NEK2A, ROCK 2, MDM2 EGF (KSP), proteasome 20S, HDAC, or survivin.
  • KSP MDM2 EGF
  • an MRD-containing antibody is administered in combination with a compound that regulates DNA replication or repair.
  • the MRD-containing antibody is administered in combination with a compound that antagonizes or reduces the activity of: BRCA1, CHK1, CHK2, E2F, E2FL, MDM2, MDM4, or PARP1.
  • an MRD-containing antibody is administered in combination with a compound that regulates transcription or translation.
  • the MRD-containing antibody is administered in combination with a compound that antagonizes or reduces the activity of IGF1R, IGF1, IGF2, PDGFRA, PDGFRB, PDGF-A, PDGF-B, PDGF-CC, PDGF-C, PDGF-D, KIT, MYC, CD28, CDK4, CDK6, mTOR, MDM2, HDAC, E2F, E2F1, or HIF1.
  • an MRD-containing antibody is administered in combination with a compound that regulates migration, invasion or metastasis.
  • the MRD-containing antibody is administered in combination with a compound that inhibits or reduces the activity of: c-MET, RON, CXCR4, PI3K, AKT, MMP2, FN1, CATHD, AMF, ⁇ v ⁇ 1 integrin, ⁇ v ⁇ 3 integrin, ⁇ v ⁇ 5 integrin, TGFb, ⁇ 1 ⁇ 1 integrin, ⁇ 2 ⁇ 1 integrin, ⁇ 4 ⁇ 1 integrin, ⁇ 2 ⁇ 1 integrin, ⁇ 5 ⁇ 1 integrin, ⁇ 9 ⁇ 1 integrin, ⁇ 6 ⁇ 4 integrin, ⁇ M ⁇ 2 integrin, or HIF1.
  • an MRD-containing antibody is administered in combination with a compound that regulates cell metabolism.
  • the MRD-containing antibody is administered in combination with a compound that inhibits or reduces the activity of: Erb132, EGFR, IGF1R, IGF1, IGF2, TGFa, ICOS, PI3K, VEGFR1, VEGFR2, mTOR, HIF1, or HDAC.
  • an MRD-containing antibody is administered in combination with an inhibitor of one or more protein kinases.
  • the protein kinase inhibitor inhibits a target of the MRD containing antibody (e.g., by either one or more MRDs or the antibody of the MRD containing antibody).
  • the protein kinase inhibitor inhibits a protein kinase that is not a target of the MRD containing antibody.
  • the protein kinase inhibitor inhibits one protein kinase.
  • the protein kinase inhibitor inhibits more than one protein kinase.
  • an MRD containing antibody is administered in combination with an inhibitor (e.g., small molecule, antibody, etc.,) of a protein kinase selected from: EGFR, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIb), FGFR3, ErbB2, VEGFR2, VEGFR3, Tie-2, PDGFR, PDGFRB, RON, and c-Met.
  • the inhibitor inhibits a protein kinase that is not targeted by the MRD containing antibody.
  • an MRD-containing antibody is administered in combination with an inhibitor of one or more protein kinases selected from: EGFR, FGFR1 (e.g., FGFR1-IIIC), FGFR2 (e.g., FGFR2-IIIa, FGFR2-IIIb, and FGFR2-IIIb), FGFR3, ErbB2, VEGFR1, VFGFR2, VEGFR3, Tie-2, PDGFRA, PDGFRB, FIT3, ALK, RET, Kit, raf, p38, RON, c-Met, PI3K, ERK, FAK, AKT, SYK, JAK1, JAK2, JAK3, TYK2, SIP, FAK, PTK7, PKD1, PKA, PKC, PKG, PRKDC, Pim, CDK, plk, p38MAPK, SRC, ABL, FGR, FYN, HCK, LCK, LYN, YES, EPH4, BMK1, ER
  • an MRD-containing antibody is administered in combination with a protein kinase inhibitor selected from: imatinib mesylate (e.g., GLEEVECTM), gefitinib (e.g., IRESSATM, Astra Zeneca), vandetanib (e.g., ZACTIMATM, Astra Zeneca), erlotinib (e.g., TARCEVATM, Genentech/OSI), sunitinib (e.g., SUTENTTM, Pfizer), lapatanib (GSK), and sorafenib (e.g., NEXAVARTM, Bayer).
  • imatinib mesylate e.g., GLEEVECTM
  • gefitinib e.g., IRESSATM, Astra Zeneca
  • vandetanib e.g., ZACTIMATM, Astra Zeneca
  • erlotinib e.g., TARCEVATM, Gene
  • an MRD-containing antibody is administered in combination with a protein kinase inhibitor selected from nilotinib (e.g., AMN107, Novartis), dasatinib (e.g., BMS 354825, BMS), ABT-869, botsutinib (e.g., SKI-606, Wyeth), cediranib, recentib, captastatin, AEE788 (Novartis), AZD0530 (AstraZeneca) Exel 7646/Exel 0999 Exelixis), cabozantinib (e.g., XL184; Exelixis), XL880/GSK1363089 (Exelixis/GSK), ARQ-197 (Arqule and Daiichi Sankyo), Inno-406 (Innovive), SGS523 (SGX), PF-2341066 (Pfizer), CI-1033 (Pfizer), motesanib (nilotini
  • an MRD-containing antibody is administered in combination with a FGFR protein kinase inhibitor selected from: sunitinib, SU5402, PD173074, TKI258 (Novartis), BIBF 1120 (Boehringer Ingelheim), brivanib (BMS-582,664), E7080 (Eisai), and TSU-68 (Taiho).
  • a FGFR protein kinase inhibitor selected from: sunitinib, SU5402, PD173074, TKI258 (Novartis), BIBF 1120 (Boehringer Ingelheim), brivanib (BMS-582,664), E7080 (Eisai), and TSU-68 (Taiho).
  • an MRD-containing antibody is administered in combination with a protein kinase inhibitor of JAK1, JAK2, JAK3, or SYK.
  • the protein kinase inhibitor is selected from: lestaurtinib, tofacitinib, SB1518, CYT387, LY3009104, TG101348, fostamatinib, BAY 61-3606, and sunitinib.
  • an ErbB2 (HER2) binding MRD-containing antibody e.g., an MRD-binding antibody that binds ErbB2 by either one or more MRDs or the antibody of the MRD containing antibody
  • HER2 (HER2) binding MRD-containing antibody is administered in combination with a protein kinase inhibitor of ErbB2.
  • a trastuzumab antibody-based MRD-containing antibody is administered in combination with a protein kinase inhibitor of ErbB2.
  • an ErbB2-binding MRD-containing antibody is administered in combination with lapatinib.
  • a trastuzumab antibody-based MRD-containing antibody is administered in combination with lapatinib.
  • an ErbB2-binding MRD-containing antibody is administered in combination with, sunitinib.
  • a trastuzumab antibody-based MRD-containing antibody is administered in combination with sunitinib.
  • an ErbB2-binding MRD-containing antibody is administered in combination with neratinib.
  • a trastuzumab antibody-based MRD-containing antibody is administered in combination with neratanib.
  • an ErbB2-binding MRD-containing antibody is administered in combination with iapatinib.
  • a trastuzumab antibody-based MRD-containing antibody is administered in combination with iapatinib.
  • an ErbB2 (HER2) binding MRD-containing antibody is administered in combination with a protein kinase inhibitor selected from canertinib (GW-572016), AV-412 (AVEO), tivozanib (AVEO), vandetanib (e.g., ZACTIMATM, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), and EKB-569 (Wyeth-Ayerst).
  • a protein kinase inhibitor selected from canertinib (GW-572016), AV-412 (AVEO), tivozanib (AVEO), vandetanib (e.g., ZACTIMATM, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), and EKB-569 (Wyeth-Ayerst
  • a trastuzumab antibody-based MRD-containing antibody is administered in combination with a protein kinase inhibitor selected from: canertinib (GW-572016), AV-412 (AVEO), tivozanib (AVEO), vandetanib (e.g., ZACTIMATM, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), PX-866 (Oncothyreon), and EKB-569 (Wyeth-Ayerst).
  • a protein kinase inhibitor selected from: canertinib (GW-572016), AV-412 (AVEO), tivozanib (AVEO), vandetanib (e.g., ZACTIMATM, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer
  • an EGFR binding MRD-containing antibody e.g., an MRD-binding antibody that binds EGFR by either one or more MRDs or the antibody of the MRD containing antibody
  • a protein kinase inhibitor of EGFR e.g., an MRD-binding antibody that binds EGFR by either one or more MRDs or the antibody of the MRD containing antibody
  • a cetuximab antibody-based MRD-containing antibody is administered in combination with a protein kinase inhibitor of EGFR.
  • an EGFR binding MRD-containing antibody is administered in combination with gefitinib (e.g., IRESSATM, AstraZeneca).
  • a cetuximab antibody-based MRD-containing antibody is administered in combination with gefitinib (e.g., IRESSATM, AstraZeneca).
  • an EGFR binding MRD-containing antibody is administered in combination with erlotinib (e.g., TARCEVATM, Genentech/OSI).
  • erlotinib e.g., TARCEVATM, Genentech/OSI
  • a cetuximab antibody-based MRD-containing antibody is administered in combination with erlotinib (e.g., TARCEVATM, Genentech/OSI).
  • an EGFR binding MRD-containing antibody is administered in combination with lapatinib.
  • a cetuximab antibody-based MRD-containing antibody is administered in combination with lapatinib.
  • an EGFR binding MRD-containing antibody is administered in combination with sorafenib (e.g., NEXAVARTM, Bayer).
  • sorafenib e.g., NEXAVARTM, Bayer
  • sorafenib e.g., NEXAVARTM, Bayer
  • an EGFR binding MRD-containing antibody is administered in combination with a protein kinase inhibitor selected from: canertinib (GW-572016), ZD6474, AV-412 (AVEO), tivozanib (AVEO), vandetanib (ZACTIMA, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), and EKB-569 (Wyeth-Ayerst).
  • a protein kinase inhibitor selected from: canertinib (GW-572016), ZD6474, AV-412 (AVEO), tivozanib (AVEO), vandetanib (ZACTIMA, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), and EKB-569 (Wyeth-Ayerst).
  • a cetuximab antibody-based MRD-containing antibody is administered in combination with a protein kinase inhibitor selected from: canertinib (GW-572016), ZD6474, AV-412 (AVEO), tivozanib (AVEO), vandetanib (ZACTIMA, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), PX-866 (Oncothyreon), and EKB-569 (Wyeth-Ayerst).
  • a protein kinase inhibitor selected from: canertinib (GW-572016), ZD6474, AV-412 (AVEO), tivozanib (AVEO), vandetanib (ZACTIMA, AstraZeneca), AEE788 (Novartis), Exel 7646/Exel 0999 (Exelixis), CI-1033 (Pfizer), PX
  • a VEGFA, VEGFR1, or VEGFR2 binding MRD-containing antibody e.g., an MRD-binding antibody that binds VEGFR1 by either one or more MRDs or the antibody of the MRD containing antibody
  • a protein kinase inhibitor of VEGR1, VEGFR2, or VEGFR3 e.g., VEGR1, VEGFR2, or VEGFR3.
  • the VEGFA, VEGFR1 or VEGFRr2 binding MRD-containing antibody is administered in combination with: sunitinib, sorafenib, pazopanib (e.g., GW786034B), AZD2171, vatalanib, ZD6474, AMG-706, or AC013736.
  • an MRD-containing antibody is administered in combination with a proteasome inhibitor.
  • the inhibitor is bortezomib (e.g., VELCADETM).
  • the inhibitor is PR-171 (Proteolix).
  • an MRD-containing antibody is administered in combination with a HDAC inhibitor.
  • an MRD-containing antibody is administered in combination with a mTOR inhibitor.
  • an MRD-containing antibody is administered in combination with a NFKB inhibitor.
  • the invention provides a method of treating cancer comprising administering a therapeutically effective amount of a VEGFA or VEGFR binding MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating cancer comprising administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient in need thereof.
  • the invention provides a method of treating colorectal cancer by administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient having colorectal cancer.
  • the invention provides a method of treating breast cancer by administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient having breast cancer.
  • the invention provides a method of treating non-small cell lung carcinoma by administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient having non-small cell lung carcinoma.
  • therapeutic effective amounts of bevacizumab comprising at least one MRD are administered to a patient to treat metastatic colorectal cancer, metastatic breast cancer, metastatic pancreatic cancer, or metastatic non-small cell lung carcinoma.
  • the invention provides a method of treating cancer by administering to a patient a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient having renal cell carcinoma, glioblastoma multiforme, ovarian cancer, prostate cancer, liver cancer or pancreatic cancer.
  • compositions of the invention are administered alone or in combination with one or more additional therapeutic agents.
  • Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially. This includes presentations in which the combined agents are administered together as a therapeutic mixture, and also procedures in which the combined agents are administered separately but simultaneously, e.g., as through separate intravenous lines into the same individual.
  • Administration “in combination” further includes the separate administration of one of the therapeutic compounds or agents given first, followed by the second.
  • a VEGFA or VEGFR binding MRD-containing antibody is administered in combination with 5-fluorouracil, carboplatin, paclitaxel, or interferon alpha.
  • bevacizumab comprising at least one MRD is administered in combination with 5-fluorouracil, carboplatin, paclitaxel, or interferon alpha.
  • the invention provides a method of treating macular degeneration comprising administering a therapeutically effective amount of a VEGFA or VEGFR binding MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating macular degeneration comprising administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient in need thereof.
  • the invention provides a method of treating macular degeneration comprising administering a therapeutically effective amount of ranibizumab comprising at least one MRD to a patient in need thereof.
  • the invention provides a method of treating cancer comprising administering a therapeutically effective amount of an ErbB2 (HER2) binding MRD-containing antibody to a patient in need thereof.
  • the ErbB2-binding multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the ErbB2-binding multivalent and multispecific compositions are administered to patients who have been previously shown to respond to another ErbB2-based therapy (e.g., HERCEPTIN, chemotherapy and/or radiation) or are predicted to respond to another ErbB2-based therapy.
  • the ErbB2-binding multivalent and multispecific compositions are administered to patients who have previously failed to respond to another ErbB2-based therapy or are predicted to fail to respond to another ErbB2-based therapy.
  • the invention provides a method of treating cancer comprising administering a therapeutically effective amount of trastuzumab comprising at least one MRD to a patient in need thereof.
  • the invention provides a method of treating breast cancer by administering a therapeutically effective amount of trastuzumab comprising at least one MRD to a patient having breast cancer.
  • therapeutic effective amounts of trastuzumab comprising at least one MRD are administered to a patient to treat metastatic breast cancer.
  • an ErbB2 (HER2) binding MRD-containing antibody is administered in combination with cyclophosphamide, paclitaxel, docetaxel, carboplatin, anthracycline, or a maytansinoid.
  • trastuzumab comprising at least one MRD is administered in combination with cyclophosphamide, paclitaxel, docetaxel, carboplatin, anthracycline, or a maytansinoid.
  • the invention provides a method of treating cancer comprising administering a therapeutically effective amount of a CD20-binding MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating a hematologic cancer comprising administering a therapeutically effective amount of rituximab comprising at least one MRD to a patient in need thereof.
  • the invention provides a method of treating CD20 positive NHL by administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient having CD20 positive NHL.
  • the invention provides a method of treating CD20 positive CLL by administering a therapeutically effective amount of bevacizumab comprising at least one MRD to a patient having CD20 positive CLL.
  • a therapeutically effective amount of a CD20-binding MRD-containing antibody is administered in combination with: ludarabine, cyclophosphamide, FC (fludarabine and cyclophosphamide), anthracycline based chemotherapy regimen (e.g., CHOP (cyclophosphamide, adriamycin, vincristine and prednisone)), or CVP (cyclophosphamide, prednisone, and vincristine) chemotherapy.
  • anthracycline based chemotherapy regimen e.g., CHOP (cyclophosphamide, adriamycin, vincristine and prednisone)
  • CVP cyclophosphamide, prednisone, and vincristine
  • a therapeutically effective amount of bevacizumab comprising at least one MRD is administered in combination with: ludarabine, cyclophosphamide, FC (fludarabine and cyclophosphamide), anthracycline based chemotherapy regimen (e.g., CHOP (cyclophosphamide, adriamycin, vincristine and prednisone)), or CVP (cyclophosphamide, prednisone, and vincristine) chemotherapy.
  • anthracycline based chemotherapy regimen e.g., CHOP (cyclophosphamide, adriamycin, vincristine and prednisone)
  • CVP cyclophosphamide, prednisone, and vincristine
  • any of the antibody-MRD fusions containing antibodies and/or MRDs that bind CD20 can be used according to the methods of treating a disorder associated with CD20, or that can be treated by targeting cells that express CD20 (e.g., hematological cancers and autoimmune disease).
  • the antibody component of the antibody-MRD-fusion is selected from rituximab, ocrelizumab, GA101, and PF-5,230,895.
  • the invention also provides a method of treating a disorder of the immune system comprising administering a therapeutically effective amount of an MRD-containing antibody.
  • the administered MRD-containing antibody binds a target selected from: CD20, TNFRSF5 (CD40), CD45RB, CD52, CD200, CCR2, PAFR, IL6R, TNFRSF1A, VLA4, CSF2, TNFSF5 (CD40 LIGAND), TLR2, TLR4, GPR44, FASL, TREM1, IL1, IL1 beta, IL1RN, tissue factor, MIF, MIP2, IL6, IL8, IL10, IL12, IL13, IL15, IL17, IL18, IL23, TNF, TNFSF12 (TWEAK), LPS, CXCL13, VEGF, IFN alpha, IFN gamma, GMCSF, FGF, TGFb, C5, and CCR3.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies
  • the invention provides a method of treating a disorder of the immune system comprising administering a therapeutically effective amount of an MRD-containing antibody that binds TNF and ANG2.
  • the invention provides a method of treating a disorder of the immune system comprising administering a therapeutically effective amount of an MRD-containing antibody that binds IL1, IL12, and TNF.
  • the MRD-containing antibody binds IL1, IL12, TNF and ANG2.
  • the administered MRD-containing antibody binds IL1, IL6 and TNF. In further embodiments, the MRD-containing antibody binds ILL IL6 TNF and ANG2.
  • target selected from: CD20, TNFRSF5 (CD40), CD45RB, CD52, CD200, CCR2, PAFR, IL6R, TNFRSF1A, VLA4, CSF2, TNFSF5 (CD40 LIGAND), TLR2, TLR4, GPR44, FASL, TREM1, IL1, IL1 beta, IL1RN, tissue factor, MIF, MIP2, IL6, IL8, IL10, IL12, IL13, IL15, IL17, IL18, IL23, TNF, TNFSF12 (TWEAK), LPS, CXCL13, VEGF, IFN alpha, IFN gamma, GMCSF, FGF, TGFb, C5, and CCR3.
  • Multivalent and multispecific compositions e.g., MRD-containing antibodies that bind 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of an MRD-containing antibody.
  • the administered MRD-containing antibody binds a target selected from: CD1C, CD3, CD4, CD19, CD20, CD21, CD22, CD23, CD24, CD28, CD37, CD38, CD45RB, CD52, CD69, CD72, CD74, CD75, CD79A, CD79B, CD80, CD81, CD83, CD86, CD200, IL2RA, IL1R2, IL6R, VLA4, HLA-DRA, HLA-A, ITGA2, ITGA3, CSF2, TLR2, TLR4, GPR44, TREM1, TIE2, TNF, FASL, tissue factor, MIF, MIP2, IL1, IL1 beta, IL1RN, IL2, IL4, IL6, IL8, IL10, IL11, IL12, IL13, IL15, IL17, IL18
  • the multivalent and multispecific compositions are administered to treat rheumatoid arthritis and the multivalent and multispecific compositions bind a target selected from: CD19, CD20, CD45RB, CD52CD200, IL1, IL6, IL12, IL15, IL17, IL18, IL23, TNF, TNFSF12 (TWEAK), TNFRSF5 (CD40), TNFSF5 (CD40 Ligand), TNFSF13B (BLyS), VEGF, VLA4, IFN gamma, IFN alpha, GMCSF, FGF, C5, CXCL13 and CCR2.
  • a target selected from: CD19, CD20, CD45RB, CD52CD200, IL1, IL6, IL12, IL15, IL17, IL18, IL23, TNF, TNFSF12 (TWEAK), TNFRSF5 (CD40), TNFSF5 (CD40 Ligand), TNFSF13B (BLyS), VEGF, VLA4, I
  • the multivalent and multispecific compositions are administered to treat systemic lupus erythematous and the multivalent and multispecific compositions bind IFN alpha and TNFSF13B (BLyS).
  • the multivalent and multispecific compositions are administered to treat multiple sclerosis and the multivalent and multispecific compositions bind a target selected from: ANG2, IL1, IL12, IL18, IL23, CXCL13, TNF, TNFRSF5 (CD40), TNFSF5 (CD40 Ligand), VEGF, VLA4, TNF, CD45RB, CD200, IFN gamma, GM-CSF, FGF, C5, CD52, TNFRSF1A, TNFRSF5 (CD40), TNFRSF6 (Fas, CD95), TNFRSF7 (CD27), TNFRSF8 (CD30), TNFSF12 (TWEAK), TNFRSF13C (BAFFR), TNFSF5 (CD40 Ligand), TNFSF6 (Fas Ligand), TNFSF8 (CD30 Ligand), TNFRSF21 (DR6), TNFSF12 (TWEAK), TNFSF13B (
  • the invention provides a method of treating, a disorder of the immune system comprising administering a therapeutically effective amount of a CD20-binding MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of a CD20-binding MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of a ritaximab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating rheumatoid arthritis comprising administering a therapeutically effective amount of a rituximab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating systemic lupus erythematous comprising administering a therapeutically effective amount of a rituximab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating multiple sclerosis comprising administering a therapeutically effective amount of a rituximab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of an ocrelizumab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating rheumatoid arthritis comprising administering a therapeutically effective amount of an ocrelizumab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating systemic lupus erythematous comprising administering a therapeutically effective amount of a ocrelizumab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating multiple sclerosis comprising administering a therapeutically effective amount of an ocrelizumab-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of a PF5,230,895-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating rheumatoid arthritis comprising administering a therapeutically effective amount of a PF5,230,895-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating systemic lupus erythematous comprising administering a therapeutically effective amount of a PF5,230,895-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating multiple sclerosis comprising administering a therapeutically effective amount of an PF5,230,895-MRD-containing antibody to a patient in need thereof.
  • the invention provides a method of treating a disorder of the immune system comprising administering a therapeutically effective amount of an MRD-containing antibody that binds CD20.
  • the administered MRD-containing antibody binds CD20 and a target selected from: TNF, TNFRSF5 (CD40), TNFSF5 (CD40 LIGAND), TNFSF12 (TWEAK), TNFRSF1A, CD45, RB, CD52, CD200, CCR2, PAFR, IL6R, VLA4, CSF2, RAGE, TLR2, TLR4, GPR44, FASL, TREM1, TIE2, tissue factor, MIF, MIP2, LPS, IL1, IL1 beta, IL1RN, IL6, IL6R, IL8, IL10, IL12, IL13, IL15, IL17, IL18, IL23, CXCL13, VEGF, IFN alpha, IFN gamma, GMCSF, FGF, C5, and CCR
  • Multivalent and multispecific compositions that bind CD20 and also bind at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds CD20.
  • the antibody component of the MRD-containing antibody is a rituximab, ocrelizumab, GA101 or PF-5,230,895.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of an MRD-containing antibody that binds CD20.
  • the administered MRD-containing antibody binds CD20 and a target selected from: CD1C, CD3, CD4, CD19, CD21, CD22, CD23, CD24, CD28, CD37, CD38, CD45RB, CD52, CD69, CD72, CD74, CD75, CD79A, CD79B, CD80, CD81, CD83, CD86, CD200, IL2RA, IL1R2, IL6R, VLA4, HLA-DRA, HLA-A, ITGA2, ITGA3, CSF2, TLR2, TLR4, GPR44, TREM1, TIE2, TNF, FASL, tissue factor, MIF, MIP2, IL1, IL1 beta, IL1RN, IL2, IL4, IL6, IL8, IL10, IL11, IL12, IL13, IL15,
  • the multivalent and multispecific compositions are administered to treat rheumatoid arthritis and the multivalent and multispecific compositions bind CD20 and a target selected from: CD19, CD45RB, CD52, CD200, IL12, IL15, IL17, IL18, IL23, TNF, TNFSF12 (TWEAK).
  • TNFRSF5 CD40
  • TNFSF5 CD40 Ligand
  • VEGF VLA4, IFN gamma, interferon alpha, GMCSF, FGF, C5, CXCL13 and CCR2.
  • the multivalent and multispecific compositions are administered to treat multiple sclerosis and the multivalent and multispecific compositions bind CD20 and a target selected from: ANG2, IL12, IL18, IL23, CXCL13, TNFRSF5 (CD40), TNFSF5 (CD40 Ligand), VEGF, VLA4, TNF, CD45RB, CD200, IFN gamma, GM-CSF, FGF, C5, CD52, TIE2, TNFRSF1A, TNFRSF5 (CD40), TNFRSF6 (Fas, CD95), TNFRSF7 (CD27), TNFRSF8 (CD30), TNFSF12 (TWEAK), TNFRSF13C (BAFFR), TNFSF5 (CD40 Ligand), TNFSF6 (Fas Ligand), TNFSF8 (CD30 Ligand), TNFRSF21 (DR6), TNFSF12 (TWEAK), TNFSF13B (
  • Multivalent and multispecific compositions that bind a least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TNF.
  • the antibody component of the MRD-containing antibody is selected from rituximab, ocrelizumab, GA101 and PF-5,230,895.
  • the invention provides a method of treating a disorder of the immune system comprising administering a therapeutically effective amount of a TNF-binding MRD-containing antibody to a patient in need thereof.
  • the TNF-binding multivalent and multispecific compositions e.g., MRD-containing antibodies
  • TNF-binding multivalent and multispecific compositions are administered to patients who have previously failed to respond to another TNF-based therapy or are predicted to fail to respond to another TNF-based therapy.
  • the invention provides a method of treating a disorder of the immune system comprising administering a therapeutically effective amount of an MRD-containing antibody that binds TNF.
  • the administered MRD-containing antibody binds TNF and a target selected from CD20, TNFRSF5 (CD40), CD45RB, CD52, CD200, CCR2, PAFR, IL6R, TNFRSF1A, VLA4, CSF2, TNFSF5 (CD40 LIGAND), TLR2, TLR4, GPR44, FASL, TREM1, IL1, IL1 beta, IL1RN, tissue factor, MIF, MIP2, IL6, IL8, IL10, IL12, IL13, IL15, IL17, IL18, IL23, TNFSF12 (TWEAK), LPS, CXCL13, VEGF, IFN gamma, GMCSF, FGF, C5, and CCR3.
  • Multivalent and multispecific compositions that bind TNF and at least 1, 2, 3, 4, 5 or more of these targets are also encompassed by the invention.
  • the antibody component of the MRD-containing antibody binds TNF.
  • the antibody component of the MRD-containing antibody is selected from adalimumab, certolizumab, golimumab and AME-527.
  • the invention provides a method of treating an autoimmune disease comprising administering a therapeutically effective amount of an MRD-containing antibody that binds TNF.
  • the administered MRD-containing antibody binds TNF and a target selected from: CD1C, CD3, CD4, CD19, CD20, CD21, CD22, CD23, CD24, CD28, CD37, CD38, CD45RB, CD52, CD69, CD72, CD74, CD75, CD79A, CD79B, CD80, CD81, CD83, CD86, CD200, IL2RA, IL1R2, IL6R, VLA4, HLA-DRA, HLA-A, ITGA2, ITGA3, CSF2, TLR2, TLR4, GPR44, TREM1, TIE2, FASL, tissue factor, MIF, MIP2, ILL IL1 beta, IL1RN, IL2, IL4, IL6, IL8, IL10, IL11, IL12, IL13, IL15, IL
  • the multivalent and multispecific compositions are administered to treat rheumatoid arthritis and the multivalent and multispecific compositions bind TNF and a target selected from CD19, CD20, CD45RB, CD52CD200, IL12, IL15, IL17, IL18, IL23, TNFSF12 (TWEAK), TNFRSF5 (CD40), TNFSF5 (CD40 Ligand), TNFSF13B (BLyS), VEGF, VLA4, IFN gamma, interferon alpha, GMCSF, FGF, C5, CXCL13 and CCR2.
US13/838,438 2011-05-24 2013-03-15 Multivalent and Monovalent Multispecific Complexes and Their Uses Abandoned US20140065142A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/838,438 US20140065142A1 (en) 2011-05-24 2013-03-15 Multivalent and Monovalent Multispecific Complexes and Their Uses
US14/481,181 US20150210765A1 (en) 2011-05-24 2014-09-09 Multivalent and Monovalent Multispecific Complexes and Their Uses
US15/458,079 US10526381B2 (en) 2011-05-24 2017-03-14 Multivalent and monovalent multispecific complexes and their uses

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161489249P 2011-05-24 2011-05-24
US201261597714P 2012-02-10 2012-02-10
US201261610831P 2012-03-14 2012-03-14
PCT/US2012/039469 WO2012162561A2 (en) 2011-05-24 2012-05-24 Multivalent and monovalent multispecific complexes and their uses
US13/838,438 US20140065142A1 (en) 2011-05-24 2013-03-15 Multivalent and Monovalent Multispecific Complexes and Their Uses

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2012/039469 Continuation WO2012162561A2 (en) 2011-05-24 2012-05-24 Multivalent and monovalent multispecific complexes and their uses
US13/838,438 Continuation US20140065142A1 (en) 2011-05-24 2013-03-15 Multivalent and Monovalent Multispecific Complexes and Their Uses

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US13/838,438 Continuation US20140065142A1 (en) 2011-05-24 2013-03-15 Multivalent and Monovalent Multispecific Complexes and Their Uses
US14/481,181 Continuation US20150210765A1 (en) 2011-05-24 2014-09-09 Multivalent and Monovalent Multispecific Complexes and Their Uses
US15/458,079 Continuation US10526381B2 (en) 2011-05-24 2017-03-14 Multivalent and monovalent multispecific complexes and their uses

Publications (1)

Publication Number Publication Date
US20140065142A1 true US20140065142A1 (en) 2014-03-06

Family

ID=46229931

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/838,438 Abandoned US20140065142A1 (en) 2011-05-24 2013-03-15 Multivalent and Monovalent Multispecific Complexes and Their Uses
US14/481,181 Abandoned US20150210765A1 (en) 2011-05-24 2014-09-09 Multivalent and Monovalent Multispecific Complexes and Their Uses
US15/458,079 Active US10526381B2 (en) 2011-05-24 2017-03-14 Multivalent and monovalent multispecific complexes and their uses

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/481,181 Abandoned US20150210765A1 (en) 2011-05-24 2014-09-09 Multivalent and Monovalent Multispecific Complexes and Their Uses
US15/458,079 Active US10526381B2 (en) 2011-05-24 2017-03-14 Multivalent and monovalent multispecific complexes and their uses

Country Status (8)

Country Link
US (3) US20140065142A1 (es)
EP (1) EP2714738B1 (es)
CN (2) CN106432506A (es)
AU (2) AU2012258637B2 (es)
CA (1) CA2837169C (es)
DK (1) DK2714738T3 (es)
ES (1) ES2704038T3 (es)
WO (1) WO2012162561A2 (es)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150274818A1 (en) * 2014-02-02 2015-10-01 Medlmmune Limited Compounds and methods for treating pain
US20150283261A1 (en) * 2011-04-04 2015-10-08 Wisconsin Alumni Research Foundation Method for selective targeting and entry of bacterial toxins to cells
WO2016070014A1 (en) * 2014-10-31 2016-05-06 H. Lee Moffitt Cancer Center And Research Institute, Inc. Tetravalent tlr9 bispecific antibody
WO2016115171A1 (en) * 2015-01-16 2016-07-21 The Regents Of The University Of California Insulin-like growth factor 2 (igf2) signaling and modulation
WO2016145385A3 (en) * 2015-03-11 2016-11-17 Integrated Biotherapeutics, Inc. Pan-ebola and pan-filovirus protective epitopes, antibodies, and antibody cocktails
US9605070B2 (en) 2014-01-31 2017-03-28 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US10077306B2 (en) * 2016-07-14 2018-09-18 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
US10150800B2 (en) 2013-03-15 2018-12-11 Zyngenia, Inc. EGFR-binding modular recognition domains
US10196445B1 (en) * 2015-03-17 2019-02-05 Bristol-Myers Squibb Company Ipilimumab variant with enhanced ADCC
US10208317B2 (en) 2013-12-11 2019-02-19 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse embryonic stem cell genome
US20190270806A1 (en) * 2018-03-02 2019-09-05 Kodiak Sciences Inc. Il-6 antibodies and fusion constructs and conjugates thereof
US10526381B2 (en) 2011-05-24 2020-01-07 Zygenia, Inc. Multivalent and monovalent multispecific complexes and their uses
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
JP2020522254A (ja) * 2017-05-31 2020-07-30 エルスター セラピューティクス, インコーポレイテッド 骨髄増殖性白血病(mpl)タンパク質に結合する多特異性分子およびその使用
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11071771B2 (en) 2014-10-17 2021-07-27 Kodiak Sciences Inc. Butyrylcholinesterase zwitterionic polymer conjugates
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
WO2022032025A1 (en) * 2020-08-05 2022-02-10 Synthekine, Inc. Ifngr binding synthetic cytokines and methods of use
EP3954703A3 (en) * 2014-05-29 2022-05-18 MacroGenics, Inc. Tri-specific binding molecules and methods of use thereof
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US11739163B2 (en) * 2016-09-29 2023-08-29 Aebi Ltd. Therapeutic multi-targeting constructs and uses thereof
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder
US11920128B2 (en) 2013-09-18 2024-03-05 Kymab Limited Methods, cells and organisms

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110189206A1 (en) 2008-01-03 2011-08-04 Barbas Iii Carlos F Antibody Targeting Through a Modular Recognition Domain
WO2012009705A1 (en) 2010-07-15 2012-01-19 Zyngenia, Inc. Ang-2 binding complexes and uses thereof
EP3549584A1 (en) 2013-03-12 2019-10-09 Stichting Het Nederlands Kanker Instituut- Antoni van Leeuwenhoek Ziekenhuis Combinations of inhibitors of mek, egfr and erbb2 in the treatment of kras-mutant lung cancer, and kras-mutant colon cancer
US9458246B2 (en) 2013-03-13 2016-10-04 Amgen Inc. Proteins specific for BAFF and B7RP1
JOP20140087B1 (ar) 2013-03-13 2021-08-17 Amgen Inc بروتينات مخصصة ل baff و b7rp1 وإستخداماتها
KR102049990B1 (ko) * 2013-03-28 2019-12-03 삼성전자주식회사 c-Met 항체 및 VEGF 결합 단편이 연결된 융합 단백질
ME03476B (me) 2013-08-01 2020-01-20 Five Prime Therapeutics Inc Nefukozilisana anti-fgfr2iiib antitijela
SG11201605093VA (en) 2013-12-23 2016-07-28 Zymeworks Inc Antibodies comprising c-terminal light chain polypeptide extensions and conjugates and methods of use thereof
CA2937521C (en) 2014-02-04 2023-08-29 Pfizer, Inc. Combination of a pd-1 antagonist and a vegfr inhibitor for treating cancer
CN106170300A (zh) * 2014-04-08 2016-11-30 西雅图基因公司 Cd19‑抗体药物偶联物的优化给药
LT3148579T (lt) 2014-05-28 2021-05-25 Agenus Inc. Anti-gitr antikūnai ir jų panaudojimo būdai
GB201409558D0 (en) 2014-05-29 2014-07-16 Ucb Biopharma Sprl Method
GB201412658D0 (en) * 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
GB201412659D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
MX2017007826A (es) * 2014-12-19 2018-02-13 Biotecnol Ltd Proteina de fusion que conprende tres dominios de union a 5t4 y cd3.
CN104774268B (zh) * 2015-01-21 2018-09-28 武汉友芝友生物制药有限公司 一种双特异性抗体egfr×cd3的构建及应用
US10800846B2 (en) 2015-02-26 2020-10-13 Merck Patent Gmbh PD-1/PD-L1 inhibitors for the treatment of cancer
CA2989586A1 (en) 2015-06-16 2016-12-22 Pfizer, Inc. Pd-l1 antagonist combination treatments
GB201601073D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies
GB201601077D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibody molecule
GB201601075D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies molecules
CA2999756A1 (en) 2015-09-24 2017-03-30 The University Of North Carolina At Chapel Hill Methods and compositions for reducing metastases
EA201890850A1 (ru) 2015-10-02 2018-09-28 Юлиус-Максимилианс-Универзитет Вюрцбург Комбинированное лечение с использованием ингибиторов человеческого фактора роста и дифференцировки 15 (gdf-15) и блокаторов контрольных точек иммунного ответа
US11262360B2 (en) 2015-10-02 2022-03-01 Julius-Maximilians-Universität Würzburg GDF-15 as a diagnostic marker to predict the clinical outcome of a treatment with immune checkpoint blockers
US11213598B2 (en) * 2015-11-12 2022-01-04 The Board Of Trustees Of The Leland Stanford Junior University Labeled probe and methods of use
WO2017091580A1 (en) * 2015-11-23 2017-06-01 Five Prime Therapeutics, Inc. Predicting response to cancer treatment with fgfr2 inhibitors
KR20180083944A (ko) 2015-12-02 2018-07-23 아게누스 인코포레이티드 항체 및 이의 사용 방법
GB201521391D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521382D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521383D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl And Ucb Celltech Method
GB201521393D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521389D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Method
CN109310756B (zh) * 2016-05-13 2022-06-28 奥美药业有限公司 新型血管生成素2,vegf双特异性拮抗剂
CN107459578B (zh) * 2016-05-31 2021-11-26 泰州迈博太科药业有限公司 一种靶向cd47与pd-l1的双功能融合蛋白
WO2018055023A1 (en) 2016-09-22 2018-03-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of lung cancer
IL265762B2 (en) 2016-10-06 2024-04-01 Merck Patent Gmbh Dosing regimen of avolumab for cancer treatment
SG11201907299XA (en) 2017-02-08 2019-09-27 Dragonfly Therapeutics Inc Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer
AU2018220736A1 (en) * 2017-02-20 2019-09-05 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
CN114989305A (zh) * 2017-03-31 2022-09-02 北京智仁美博生物科技有限公司 新型双特异性抗体及其用途
CA3061505A1 (en) 2017-04-24 2018-11-01 Ohio State Innovation Foundation Recombinant egfl7, egfl7 antibodies, and uses thereof
CN110621336A (zh) * 2017-05-16 2019-12-27 戊瑞治疗有限公司 癌症治疗中抗fgfr2抗体与化学治疗剂的组合
AU2018271872A1 (en) * 2017-05-23 2019-12-12 Dragonfly Therapeutics, Inc. A protein binding NKG2D, CD16 and ROR1 or ROR2
EP3749346A4 (en) 2018-02-08 2021-12-15 Dragonfly Therapeutics, Inc. VARIABLE DOMAINS OF ANTIBODIES TARGETING THE NKG2D RECEIVER
WO2019197605A2 (en) 2018-04-12 2019-10-17 Umc Utrecht Holding B.V. Drug combinations for use in the treatment of ras-mutant cancer
EA202091888A1 (ru) * 2018-08-08 2020-10-23 Драгонфлай Терапьютикс, Инк. Вариабельные домены антител, нацеленные на рецептор nkg2d
JP2022525238A (ja) 2019-04-01 2022-05-11 インメタス セラピューティクス, インコーポレイテッド 腫瘍微小環境および免疫チェックポイントタンパク質を標的とする二重特異性結合分子
BR112022001273A2 (pt) * 2019-08-02 2022-06-07 Univ California Composições e métodos para direcionar e matar células tronco cancerosas (cscs) positivas para alfa-v beta-3 (avss3) (cscs) e tratar cânceres metastáticos e resistentes a fármacos
EP4210702A1 (en) 2020-09-14 2023-07-19 JANSSEN Pharmaceutica NV Fgfr inhibitor combination therapies
WO2023102875A1 (en) * 2021-12-10 2023-06-15 Multitude Therapeutics Inc. Anti-cdh6 antibody drug conjugate

Family Cites Families (316)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CU22545A1 (es) 1994-11-18 1999-03-31 Centro Inmunologia Molecular Obtención de un anticuerpo quimérico y humanizado contra el receptor del factor de crecimiento epidérmico para uso diagnóstico y terapéutico
FR2413974A1 (fr) 1978-01-06 1979-08-03 David Bernard Sechoir pour feuilles imprimees par serigraphie
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
US4714681A (en) 1981-07-01 1987-12-22 The Board Of Reagents, The University Of Texas System Cancer Center Quadroma cells and trioma cells and methods for the production of same
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4599304A (en) * 1983-10-07 1986-07-08 Becton, Dickinson And Company Method for monitoring activated cell subpopulations
US6054561A (en) 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
JPS6147500A (ja) 1984-08-15 1986-03-07 Res Dev Corp Of Japan キメラモノクロ−ナル抗体及びその製造法
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
JP2532858B2 (ja) 1985-04-01 1996-09-11 セルテツク リミテツド 形質転換したミエロ―マ細胞系
US5374548A (en) 1986-05-02 1994-12-20 Genentech, Inc. Methods and compositions for the attachment of proteins to liposomes using a glycophospholipid anchor
MX9203291A (es) 1985-06-26 1992-08-01 Liposome Co Inc Metodo para acoplamiento de liposomas.
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
DE3689123T2 (de) 1985-11-01 1994-03-03 Xoma Corp Modulare einheit von antikörpergenen, daraus hergestellte antikörper und verwendung.
US5618920A (en) 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6121424A (en) 1991-11-25 2000-09-19 Enzon, Inc. Multivalent antigen-binding proteins
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
GB8705477D0 (en) 1987-03-09 1987-04-15 Carlton Med Prod Drug delivery systems
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
US5720937A (en) 1988-01-12 1998-02-24 Genentech, Inc. In vivo tumor detection assay
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
US4925648A (en) 1988-07-29 1990-05-15 Immunomedics, Inc. Detection and treatment of infectious and inflammatory lesions
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
CA2006408A1 (en) * 1988-12-27 1990-06-27 Susumu Iwasa Bispecific monoclonal antibody, its production and use
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DK0479909T3 (da) 1989-06-29 1997-04-07 Medarex Inc Bispecifikke reagenser til AIDS-behandling
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
CA2026147C (en) 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5196193A (en) 1989-10-31 1993-03-23 Ophidian Pharmaceuticals, Inc. Antivenoms and methods for making antivenoms
EP1690935A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
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
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
ES2246502T3 (es) 1990-08-29 2006-02-16 Genpharm International, Inc. Animales no humanos transgenicos capaces de producir anticuerpos heterologos.
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
ATE175118T1 (de) 1990-10-05 1999-01-15 Medarex Inc Gezielte immunostimulierung mit bispezifischen stoffen
EP0557300B1 (en) 1990-10-29 1997-11-19 Chiron Corporation Bispecific antibodies, method of production, and uses thereof
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
CA2082160C (en) 1991-03-06 2003-05-06 Mary M. Bendig Humanised and chimeric monoclonal antibodies
IE921342A1 (en) 1991-04-26 1992-11-04 Surface Active Ltd Novel antibodies, and methods for their use
JPH06507398A (ja) 1991-05-14 1994-08-25 リプリジェン コーポレーション Hiv感染治療のための異種複合抗体
LU91067I2 (fr) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab et ses variantes et dérivés immuno chimiques y compris les immotoxines
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
US5637481A (en) 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US5733731A (en) 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
US5270170A (en) 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
ATE275198T1 (de) 1991-12-02 2004-09-15 Medical Res Council Herstellung von antikörpern auf phagenoberflächen ausgehend von antikörpersegmentbibliotheken.
US6004554A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for targeting the vasculature of solid tumors
US5877289A (en) 1992-03-05 1999-03-02 The Scripps Research Institute Tissue factor compositions and ligands for the specific coagulation of vasculature
US5965132A (en) 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
DE69334191T2 (de) 1992-03-13 2008-11-27 bioMérieux B.V. Epstein-Barr-Virus verwandte Peptide und Nukleinsäuresegmente
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
CA2118508A1 (en) 1992-04-24 1993-11-11 Elizabeth S. Ward Recombinant production of immunoglobulin-like domains in prokaryotic cells
ES2136664T3 (es) 1992-06-09 1999-12-01 Hoppe Ag Sistema de cierre y conjunto de cerradura.
US5681714A (en) 1992-07-30 1997-10-28 Mount Sinai Hospital Corporation Nucleic acid encoding tek receptor tyrosine kinase
ATE149570T1 (de) 1992-08-17 1997-03-15 Genentech Inc Bispezifische immunoadhesine
US5844094A (en) 1992-09-25 1998-12-01 Commonwealth Scientific And Industrial Research Organization Target binding polypeptide
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US6955900B1 (en) 1993-02-02 2005-10-18 The Scripps Research Institute Methods for producing polypeptide binding sites, monoclonal antibodies and compositions thereof
US6733752B1 (en) 1994-03-30 2004-05-11 The Trustees Of The University Of Pennsylvania Prevention of tumors with monoclonal antibodies against neu
US5885573A (en) 1993-06-01 1999-03-23 Arch Development Corporation Methods and materials for modulation of the immunosuppressive activity and toxicity of monoclonal antibodies
AU691811B2 (en) 1993-06-16 1998-05-28 Celltech Therapeutics Limited Antibodies
US5922545A (en) 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
SE9400088D0 (sv) 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
US5643755A (en) 1994-10-07 1997-07-01 Regeneron Pharmaceuticals Inc. Nucleic acid encoding tie-2 ligand
US5879672A (en) 1994-10-07 1999-03-09 Regeneron Pharmaceuticals, Inc. Tie-2 ligand 1
GB9401182D0 (en) 1994-01-21 1994-03-16 Inst Of Cancer The Research Antibodies to EGF receptor and their antitumour effect
AUPM379494A0 (en) 1994-02-10 1994-03-03 Ludwig Institute For Cancer Research Immunointeractive molecules - ii
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
WO1995024220A1 (en) 1994-03-07 1995-09-14 Medarex, Inc. Bispecific molecules having clinical utilities
US5733757A (en) 1995-12-15 1998-03-31 The Scripps Research Institute Aldolase catalytic antibody
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
DE69534530T2 (de) 1994-08-12 2006-07-06 Immunomedics, Inc. Für b-zell-lymphom und leukämiezellen spezifische immunkonjugate und humane antikörper
ES2297832T3 (es) 1994-08-19 2008-05-01 La Region Wallonne Conjugados que comprenden un agente antitumoral y su utilizacion.
US5814464A (en) 1994-10-07 1998-09-29 Regeneron Pharma Nucleic acids encoding TIE-2 ligand-2
US7063840B2 (en) 1994-10-07 2006-06-20 Regeneron Pharmaceuticals, Inc. TIE-2 ligands, methods of making and uses thereof
US5650490A (en) 1994-10-07 1997-07-22 Regeneron Pharmaceuticals, Inc. Tie-2 ligand 2
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
AU715621B2 (en) 1995-04-06 2000-02-03 Regeneron Pharmaceuticals, Inc. TIE-2 ligands, methods of making and uses thereof
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
EP1709970A1 (en) 1995-04-27 2006-10-11 Abgenix, Inc. Human antibodies against EGFR, derived from immunized xenomice
CA2219486A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
JPH11507535A (ja) 1995-06-07 1999-07-06 イムクローン システムズ インコーポレイテッド 腫瘍の成長を抑制する抗体および抗体フラグメント類
AU6113396A (en) 1995-06-14 1997-01-15 Regents Of The University Of California, The Novel high affinity human antibodies to tumor antigens
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
AU716785B2 (en) 1995-07-27 2000-03-09 Genentech Inc. Stabile isotonic lyophilized protein formulation
US6828422B1 (en) 1995-08-18 2004-12-07 Morphosys Ag Protein/(poly)peptide libraries
ATE219517T1 (de) 1995-08-18 2002-07-15 Morphosys Ag Protein-/(poly)peptidbibliotheken
JP2002514895A (ja) 1995-09-28 2002-05-21 アレクション、ファーマスーティカルズ、インコーポレーテッド ブタ細胞相互作用タンパク質
US5783186A (en) 1995-12-05 1998-07-21 Amgen Inc. Antibody-induced apoptosis
HU221984B1 (hu) 1996-02-09 2003-03-28 Basf Ag Humán TNFalfa-t kötő antitestek, ezeket tartalmazó gyógyszerkészítmények és alkalmazásuk
US6194177B1 (en) 1996-02-20 2001-02-27 Applied Research Systems Ars Holding N.V. DNA encoding a hybrid heterodimeric protein
US7063965B2 (en) 1996-04-05 2006-06-20 Regeneron Pharmaceuticals, Inc. Nucleic acid encoding TIE-2 ligand
US20030040463A1 (en) 1996-04-05 2003-02-27 Wiegand Stanley J. TIE-2 ligands, methods of making and uses thereof
US6265564B1 (en) 1996-08-02 2001-07-24 Regeneron Pharmaceuticals, Inc. Expressed ligand-vascular intercellular signalling molecule
ATE230850T1 (de) 1996-10-08 2003-01-15 Bisys B V U Verfahren und mittel zur auswahl von peptiden und proteinen mit spezifischer affinität zu einem zielmolekül
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
WO1998018914A1 (en) 1996-10-31 1998-05-07 Duke University Soluble tie2 receptor
CA2722378C (en) 1996-12-03 2015-02-03 Amgen Fremont Inc. Human antibodies that bind tnf.alpha.
WO1998033914A1 (en) 1997-01-31 1998-08-06 University Of Rochester Chimeric antibody fusion proteins for the recruitment and stimulation of an antitumor immune response
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US6884879B1 (en) 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
IL132560A0 (en) 1997-05-02 2001-03-19 Genentech Inc A method for making multispecific antibodies having heteromultimeric and common components
US20030207346A1 (en) 1997-05-02 2003-11-06 William R. Arathoon Method for making multispecific antibodies having heteromultimeric and common components
US20020062010A1 (en) 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
DE69833755T2 (de) 1997-05-21 2006-12-28 Biovation Ltd. Verfahren zur herstellung von nicht-immunogenen proteinen
US6207805B1 (en) 1997-07-18 2001-03-27 University Of Iowa Research Foundation Prostate cell surface antigen-specific antibodies
WO1999004705A1 (en) 1997-07-25 1999-02-04 Tsui Ban C H Devices, systems and methods for determining proper placement of epidural catheters
US6417168B1 (en) 1998-03-04 2002-07-09 The Trustees Of The University Of Pennsylvania Compositions and methods of treating tumors
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
ATE244758T1 (de) 1998-04-21 2003-07-15 Micromet Ag Cd19xcd3 spezifische polypeptide und deren verwendung
PT1308456E (pt) 1998-05-06 2007-12-03 Genentech Inc Purificação de anticorpos por cromatografia de permuta iónica
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
US6818213B1 (en) * 1998-07-13 2004-11-16 Board Of Regents, The University Of Texas System Cancer treatment compositions comprising therapeutic conjugates that bind to aminophospholipids
US6875741B2 (en) 1998-09-02 2005-04-05 Renuka Pillutla Insulin and IGF-1 receptor agonists and antagonists
DK1113810T3 (da) 1998-09-14 2009-04-06 Univ Texas Fremgangsmåder til behandling af multipelt myelom og myelom-induceret knogleresorption under anvendelse af antagonister af integrin/receptor-binding
US6365154B1 (en) 1998-09-28 2002-04-02 Smithkline Beecham Corporation Tie2 agonist antibodies
US6376653B1 (en) 1998-09-28 2002-04-23 Smithkline Beecham Plc Tie2 antagonist antibodies
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
AU776910B2 (en) 1998-12-08 2004-09-23 Merck Patent Gesellschaft Mit Beschrankter Haftung Modifying protein immunogenicity
WO2000034337A1 (en) 1998-12-10 2000-06-15 Tsukuba Research Laboratory, Toagosei Co., Ltd. Humanized monoclonal antibodies against vascular endothelial cell growth factor
US7425541B2 (en) 1998-12-11 2008-09-16 Medarex, Inc. Enzyme-cleavable prodrug compounds
PL199645B1 (pl) 1998-12-23 2008-10-31 Regeneron Pharma Kwas nukleinowy, polipeptyd fuzyjny, kompozycja, wektor, wektor ekspresyjny, układ gospodarz wektor, sposób wytwarzania polipeptydu fuzyjnego, kwas nukleinowy i sposób wytwarzania polipeptydu fuzyjnego
EP2364997A3 (en) 1999-01-15 2012-07-04 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6897044B1 (en) 1999-01-28 2005-05-24 Biogen Idec, Inc. Production of tetravalent antibodies
PL200919B1 (pl) 1999-02-12 2009-02-27 Lexigen Pharm Corp Zastosowanie kompozycji czynnika hamującego angiogenezę i czynnika przeciwnowotworowego, kompozycja terapeutyczna do leczenia nowotworów i zestaw do traktowania komórki nowotworowej
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
US6455035B1 (en) 1999-03-26 2002-09-24 Regeneron Pharmaceuticals, Inc. Angiopoietins and methods of use thereof
EP1046715A1 (en) 1999-04-23 2000-10-25 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Interaction of vascular-endothelial protein-tyrosine phosphatase with the angiopoietin receptor tie-2
US7527787B2 (en) 2005-10-19 2009-05-05 Ibc Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US6268488B1 (en) 1999-05-25 2001-07-31 Barbas, Iii Carlos F. Prodrug activation using catalytic antibodies
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
US6521424B2 (en) 1999-06-07 2003-02-18 Immunex Corporation Recombinant expression of Tek antagonists
JP4587626B2 (ja) 1999-06-07 2010-11-24 イミュネックス・コーポレーション Tekアンタゴニスト
HU226742B1 (en) 1999-06-25 2009-08-28 Genentech Inc Humanized anti-erbb2 antibodies and treatment with anti-erbb2 antibodies
US6949245B1 (en) 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
WO2001000238A1 (en) 1999-06-25 2001-01-04 Genentech, Inc. TREATING PROSTATE CANCER WITH ANTI-ErbB2 ANTIBODIES
DE60044997D1 (de) 1999-07-02 2010-11-04 Morphosys Ag Erzeugung spezifischer bindungspartner die an von genomischen dns-fragmenten oder ests kodierten (poly)peptiden binden
NZ517150A (en) 1999-08-27 2005-01-28 Genentech Inc Dosages for treatment with anti-ErbB2 antibodies
US6497909B1 (en) 1999-09-09 2002-12-24 General Mills, Inc. Method of bleaching cereal grain
US20020173627A1 (en) 1999-09-16 2002-11-21 Samuel Davis TIE-2 ligands, methods of making and uses thereof
US20040001827A1 (en) 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
US20050287153A1 (en) 2002-06-28 2005-12-29 Genentech, Inc. Serum albumin binding peptides for tumor targeting
AU4314801A (en) 2000-02-11 2001-08-20 Lexigen Pharm Corp Enhancing the circulating half-life of antibody-based fusion proteins
US20040191260A1 (en) 2003-03-26 2004-09-30 Technion Research & Development Foundation Ltd. Compositions capable of specifically binding particular human antigen presenting molecule/pathogen-derived antigen complexes and uses thereof
LT2857516T (lt) 2000-04-11 2017-09-11 Genentech, Inc. Multivalentiniai antikūnai ir jų panaudojimas
JP2003530871A (ja) 2000-04-21 2003-10-21 アムジエン・インコーポレーテツド インテグリン/接着因子アンタゴニスト
PL400669A1 (pl) 2000-05-19 2012-11-05 Genentech, Inc. Genowa próba wykrywania odpowiedzi na antagoniste ErbB dla poprawienia prawdopodobienstwa i skutecznosci leczenia raka
US6725230B2 (en) 2000-07-18 2004-04-20 Aegis Analytical Corporation System, method and computer program for assembling process data of multi-database origins using a hierarchical display
AU8822601A (en) 2000-07-31 2002-02-13 Biolex Inc Expression of biologically active polypeptides in duckweed
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US7105348B2 (en) 2000-10-31 2006-09-12 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
ES2727425T3 (es) 2000-12-12 2019-10-16 Medimmune Llc Moléculas con semividas prolongadas, composiciones y usos de las mismas
KR100983997B1 (ko) 2001-01-09 2010-09-28 메르크 파텐트 게엠베하 수용체 타이로신 키나아제 저해제 및 혈관형성 저해제를 사용하는 병용 요법
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
NZ527591A (en) 2001-01-17 2006-04-28 Trubion Pharmaceuticals Inc Binding domain-immunoglobulin fusion proteins
US7319139B2 (en) 2001-01-29 2008-01-15 Biogen Idec, Inc. TAG-72 specific CH2 domain deleted antibodies
KR20040074587A (ko) 2001-02-19 2004-08-25 메르크 파텐트 게엠베하 감소된 면역원성을 갖는 인공 단백질
KR100900176B1 (ko) 2001-03-07 2009-06-02 메르크 파텐트 게엠베하 하이브리드 이소타입 항체 부분구조를 포함하는 단백질을위한 발현 기술
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
IL145035A0 (en) 2001-08-21 2002-06-30 Yeda Res & Dev Molecular linkers suitable for crystallization and structural analysis of molecules of interest and method of using same
US7138370B2 (en) 2001-10-11 2006-11-21 Amgen Inc. Specific binding agents of human angiopoietin-2
US7205275B2 (en) 2001-10-11 2007-04-17 Amgen Inc. Methods of treatment using specific binding agents of human angiopoietin-2
US7521053B2 (en) 2001-10-11 2009-04-21 Amgen Inc. Angiopoietin-2 specific binding agents
US7658924B2 (en) 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
ES2402527T3 (es) 2001-12-27 2013-05-06 Glycofi, Inc. Procedimientos para obtener estructuras de carbohidrato de tipo mamífero mediante ingeniería genética
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US7897842B2 (en) 2002-03-19 2011-03-01 Plant Research International B.V. GnTIII expression in plants
US7736652B2 (en) 2002-03-21 2010-06-15 The Regents Of The University Of California Antibody fusion proteins: effective adjuvants of protein vaccination
US20070003514A1 (en) 2002-04-05 2007-01-04 The Regents Of The University Of California Mono-and bi-functional antibody conjugates as effective adjuvants of protein vaccination
US7332585B2 (en) 2002-04-05 2008-02-19 The Regents Of The California University Bispecific single chain Fv antibody molecules and methods of use thereof
US20070104710A1 (en) 2002-06-28 2007-05-10 Domants Limited Ligand that has binding specificity for IL-4 and/or IL-13
SI1517921T1 (sl) 2002-06-28 2006-10-31 Domantis Ltd Dvojno-specificni ligandi z zvisano serumsko razpolovno dobo
JP5069843B2 (ja) 2002-07-15 2012-11-07 ジェネンテック, インコーポレイテッド 抗ErbB2抗体を用いる処置に応答性である腫瘍を同定するための方法
WO2006028429A2 (en) 2002-08-05 2006-03-16 The Johns Hopkins University Peptides for targeting the prostate specific membrane antigen
US7425620B2 (en) 2002-08-14 2008-09-16 Scott Koenig FcγRIIB-specific antibodies and methods of use thereof
DK1539966T3 (da) 2002-09-12 2010-10-04 Greenovation Biotech Gmbh Fremgangsmåde til fremstilling af proteiner
US20040057969A1 (en) 2002-09-20 2004-03-25 Smith Mark L Compositions containing stabilized hepatitis antigen and methods of their use
US9809654B2 (en) 2002-09-27 2017-11-07 Vaccinex, Inc. Targeted CD1d molecules
CN1705491B (zh) 2002-09-27 2013-05-29 赞科股份有限公司 优化的Fc变体及其产生方法
US7541440B2 (en) 2002-09-30 2009-06-02 Immunomedics, Inc. Chimeric, human and humanized anti-granulocyte antibodies and methods of use
JP4790413B2 (ja) 2002-10-08 2011-10-12 イミューノメディクス、インコーポレイテッド 抗体療法
KR20050057631A (ko) 2002-10-10 2005-06-16 메르크 파텐트 게엠베하 2특이적 항-erb-b 항체 및 종양 치료법에서의 이들의용도
AU2003286467B2 (en) 2002-10-15 2009-10-01 Abbvie Biotherapeutics Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
CA2505991C (en) 2002-11-15 2018-02-27 Genmab A/S Human monoclonal antibodies against cd25
JP4559358B2 (ja) 2002-12-20 2010-10-06 グリーンオベーション バイオテク ゲーエムベーハー コケ細胞における異種グリコシル化タンパク質の産生
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
PL222222B1 (pl) 2003-01-22 2016-07-29 Glycart Biotechnology Ag Sposób wytwarzania polipeptydu
JP4532409B2 (ja) 2003-01-23 2010-08-25 小野薬品工業株式会社 ヒトpd−1に対し特異性を有する物質
WO2004076650A2 (en) 2003-02-27 2004-09-10 The Trustees Of The University Of Pennsylvania Angiopoietin and fragments, mutants, and analogs thereof and uses of the same
US6902962B2 (en) 2003-04-04 2005-06-07 Taiwan Semiconductor Manufacturing Company, Ltd. Silicon-on-insulator chip with multiple crystal orientations
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
DK1629011T3 (da) 2003-05-31 2010-05-03 Micromet Ag Humane anti-humane-DC3-bindingsmolekyler
WO2005012493A2 (en) 2003-07-31 2005-02-10 Immunomedics, Inc. Anti-cd19 antibodies
US7871610B2 (en) 2003-08-12 2011-01-18 Dyax Corp. Antibodies to Tie1 ectodomain
US7485297B2 (en) 2003-08-12 2009-02-03 Dyax Corp. Method of inhibition of vascular development using an antibody
GB0320878D0 (en) 2003-09-05 2003-10-08 Celltech R&D Ltd A protein involved in carcinoma
GB0324368D0 (en) 2003-10-17 2003-11-19 Univ Cambridge Tech Polypeptides including modified constant regions
BR122018071808B8 (pt) 2003-11-06 2020-06-30 Seattle Genetics Inc conjugado
US7968684B2 (en) 2003-11-12 2011-06-28 Abbott Laboratories IL-18 binding proteins
US20050136044A1 (en) 2003-12-04 2005-06-23 Watkins Jeffry D. Butyrylcholinesterase variants that alter the activity of chemotherapeutic agents
WO2005070966A2 (en) 2004-01-16 2005-08-04 Regeneron Pharmaceuticals, Inc. Fusion polypeptides capable of activating receptors
CA2559719A1 (en) 2004-03-17 2005-09-29 University Of Hawaii Sensor constructs and detection methods
US7598350B2 (en) 2004-03-19 2009-10-06 Imclone Llc Human anti-epidermal growth factor receptor antibody
ATE537189T1 (de) 2004-04-30 2011-12-15 Inst Nat Sante Rech Med Anti-tfr-antikörper
EP1778270A4 (en) 2004-05-05 2009-09-02 Merrimack Pharmaceuticals Inc BIS-SPECIFIC BINDER FOR MODULATING BIOLOGICAL ACTIVITY
EP1778264A2 (en) 2004-06-25 2007-05-02 Licentia, Ltd. Tie receptor and tie ligand materials and methods for modulating female fertility
WO2006005361A1 (en) 2004-07-13 2006-01-19 Centre National De La Recherche Scientifique Peptides liable to bind to the tie2 receptor, nucleic acids encoding them, and their uses
DK1776384T3 (da) 2004-08-04 2013-09-02 Mentrik Biotech Llc VARIANT-Fc-REGIONER
JP2008532476A (ja) 2004-08-12 2008-08-21 ダイアックス コーポレーション 複合結合タンパク質
WO2006028936A2 (en) 2004-09-02 2006-03-16 Genentech, Inc. Heteromultimeric molecules
RU2412947C2 (ru) 2004-09-23 2011-02-27 Дженентек, Инк. Антитела, сконструированные на основе цистеинов, и их конъюгаты
SI1797127T1 (sl) 2004-09-24 2017-09-29 Amgen Inc. Modificirane fc-molekule
US20060222653A1 (en) 2004-11-12 2006-10-05 Xencor, Inc. Antibodies operably linked to selected chemoattractants
CA2587143C (en) 2004-12-08 2017-12-05 Immunomedics, Inc. Methods and compositions for immunotherapy and detection of inflammatory and immune-dysregulatory disease, infectious disease, pathologic angiogenesis and cancer
PT1838733E (pt) 2004-12-21 2011-12-13 Medimmune Ltd Anticorpos direccionados a angiopoietina-2 e respectivos usos
CA2594356C (en) 2005-01-05 2018-07-17 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H. Synthetic immunoglobulin domains with binding properties engineered in regions of the molecule different from the complementarity determining regions
KR20190110637A (ko) 2005-01-21 2019-09-30 제넨테크, 인크. Her 항체의 고정 용량 투여법
US7700099B2 (en) 2005-02-14 2010-04-20 Merck & Co., Inc. Non-immunostimulatory antibody and compositions containing the same
AU2005327973A1 (en) 2005-02-23 2006-08-31 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
KR101374454B1 (ko) 2005-03-31 2014-03-17 추가이 세이야쿠 가부시키가이샤 회합제어에 의한 폴리펩티드 제조방법
EP1896503B1 (en) 2005-05-31 2014-10-29 Board of Regents, The University of Texas System IgG1 ANTIBODIES WITH MUTATED Fc PORTION FOR INCREASED BINDING TO FcRn RECEPTOR AND USES TEHEREOF
NZ623901A (en) 2005-08-03 2015-10-30 Immunogen Inc Immunoconjugate formulations
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
WO2007024715A2 (en) 2005-08-19 2007-03-01 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US7438381B2 (en) 2005-10-11 2008-10-21 Silverbrook Research Pty Ltd Method of removing flooded ink from a printhead
JP2009514537A (ja) 2005-11-03 2009-04-09 ジェネンテック・インコーポレーテッド 治療的抗her2抗体融合ポリペプチド
CA2627190A1 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
EP1790358A1 (en) 2005-11-23 2007-05-30 Université de Reims Champagne-Ardennes Protein constructs designed for targeting and lysis of cells
GB0524788D0 (en) 2005-12-05 2006-01-11 Affibody Ab Polypeptides
KR20080077261A (ko) 2005-12-06 2008-08-21 도만티스 리미티드 Egfr 및/또는 vegf에 대해 결합 특이성이 있는리간드 및 그의 사용 방법
BRPI0619460A2 (pt) 2005-12-06 2011-11-08 Domantis Ltd ligando, uso do ligando, composição, dispositivo de dispensação de medicamento, ácido nucléico isolado ou recombinante, vetor, célula hospedeira, e, método para produzir um ligando
BRPI0619853A2 (pt) 2005-12-15 2016-08-23 Astrazeneca Ab combinação, composição farmacêutica, e, métodos de antagonização da atividade biológica, de tratamento de angiogênese relacionada à doença em um mamífero e de tratamento de câncer em um mamífero
CN101374546B (zh) 2005-12-16 2016-03-23 Ibc医药公司 基于免疫球蛋白的多价生物活性装配体
AR059066A1 (es) 2006-01-27 2008-03-12 Amgen Inc Combinaciones del inhibidor de la angiopoyetina -2 (ang2) y el inhibidor del factor de crecimiento endotelial vascular (vegf)
RU2008135433A (ru) 2006-02-02 2010-03-10 Дзе Дженерал Хоспитал Корпорейшн (Us) Гибриды рекомбинантных антител-белков стресса
EP1820513A1 (en) 2006-02-15 2007-08-22 Trion Pharma Gmbh Destruction of tumor cells expressing low to medium levels of tumor associated target antigens by trifunctional bispecific antibodies
CA2643144A1 (en) 2006-02-24 2007-08-30 Arius Research, Inc. Cancerous disease modifying antibody 141205-04
CA3022097C (en) 2006-03-15 2020-10-27 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
BRPI0710826A2 (pt) 2006-04-21 2011-08-23 Novartis Ag composições farmacêuticas de anticorpo anti-cd40 antagonista
US20100021379A1 (en) 2006-06-29 2010-01-28 The Regents Of The University Of California Chemical Antibodies for Immunotherapy and Imaging
AT503889B1 (de) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F Multivalente immunglobuline
DK2511301T3 (en) 2006-08-04 2018-03-12 Medimmune Ltd HUMAN ANTIBODIES AGAINST ERBB 2
JP5959795B2 (ja) * 2006-08-18 2016-08-02 アーメイゲン・テクノロジーズ・インコーポレイテッドArmagen Technologies, Inc. 血液脳関門送達のための物質
US20100297103A1 (en) 2006-09-14 2010-11-25 Medical & Biological Laboratories Co., Ltd. Antibody having enhanced adcc activity and method for production thereof
LT2769992T (lt) 2006-10-02 2021-04-12 Regeneron Pharmaceuticals, Inc. Didelio afiniškumo žmogaus antikūnai, atpažįstantys žmogaus il-4 receptorių
DK2076290T3 (en) 2006-10-27 2017-01-23 Sunnybrook Health Sciences Center MULTIMERIC TIE 2 AGONISTS AND APPLICATIONS THEREOF IN STIMULATING ANGIOGENESIS
WO2008070042A2 (en) 2006-12-04 2008-06-12 Medimmune, Inc. High potency recombinant antibodies, methods for producing them and use in cancer therapy
TW200831538A (en) 2006-12-19 2008-08-01 Genentech Inc VEGF-specific antagonists for adjuvant and neoadjuvant therapy and the treatment of early stage tumors
JP2010516675A (ja) 2007-01-17 2010-05-20 イミューノメディクス、インコーポレイテッド 治療薬剤のポリマー担体および疾病部位の抗体に基づく標的化のための認識部分
EP2114985B1 (en) 2007-02-02 2014-12-17 Baylor Research Institute Multivariable antigens complexed with targeting humanized monoclonal antibody
US10259860B2 (en) 2007-02-27 2019-04-16 Aprogen Inc. Fusion proteins binding to VEGF and angiopoietin
PE20090681A1 (es) 2007-03-02 2009-06-10 Genentech Inc Prediccion de respuesta a un inhibidor her
GB2447689B (en) 2007-03-22 2011-08-24 Knorr Bremse Systeme Trailer electronic braking system
JP2010521526A (ja) 2007-03-23 2010-06-24 ザ・ガバナーズ・オブ・ザ・ユニバーシティー・オブ・アルバータ 多価ヘテロ二官能性ポリマーおよびその使用方法
JP2010535032A (ja) 2007-07-31 2010-11-18 メディミューン,エルエルシー 多重特異性エピトープ結合性タンパク質およびその用途
KR101248422B1 (ko) 2007-11-13 2013-04-09 테바 바이오파머수티컬스 유에스에이, 아이엔씨. Tl1a에 대한 인간화된 항체
EP2615115A3 (en) 2007-11-30 2014-01-08 Glaxo Group Limited Antigen-binding constructs
US20090162359A1 (en) * 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US20110189206A1 (en) 2008-01-03 2011-08-04 Barbas Iii Carlos F Antibody Targeting Through a Modular Recognition Domain
US8454960B2 (en) 2008-01-03 2013-06-04 The Scripps Research Institute Multispecific antibody targeting and multivalency through modular recognition domains
US8574577B2 (en) 2008-01-03 2013-11-05 The Scripps Research Institute VEGF antibodies comprising modular recognition domains
US8557242B2 (en) 2008-01-03 2013-10-15 The Scripps Research Institute ERBB2 antibodies comprising modular recognition domains
US20140127200A1 (en) 2008-01-03 2014-05-08 The Scripps Research Institute Multispecific Antibody Targeting and Multivalency Through Modular Recognition Domains
US8557243B2 (en) 2008-01-03 2013-10-15 The Scripps Research Institute EFGR antibodies comprising modular recognition domains
CA2712000A1 (en) 2008-01-28 2009-08-06 Medimmune Limited Stabilized angiopoietin-2 antibodies and uses thereof
JO2913B1 (en) 2008-02-20 2015-09-15 امجين إنك, Antibodies directed towards angiopoietin-1 and angiopoietin-2 proteins and their uses
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
US20110076723A1 (en) 2008-05-23 2011-03-31 Samsung Electronics Co., Ltd. Antibody-peptide fused synergibody
EP2671891A3 (en) 2008-06-27 2014-03-05 Amgen Inc. Ang-2 inhibition to treat multiple sclerosis
AU2009275135A1 (en) 2008-07-21 2010-01-28 Compugen Ltd. Angiopoietin derived peptides
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
US8858940B2 (en) 2008-12-10 2014-10-14 Ablynx N.V. Amino acid sequences directed against the Angiopoietin/Tie system and polypeptides comprising the same for the treatment of diseases and disorders related to angiogenesis
US8133979B2 (en) 2008-12-16 2012-03-13 Hoffmann-La Roche Inc. Antibodies against human angiopoietin 2
EP2396036B1 (en) 2009-02-13 2017-06-28 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20120121591A1 (en) 2009-03-20 2012-05-17 Amgen Inc. SELECTIVE AND POTENT PEPTIDE INHIBITORS OF Kv1.3
US8703132B2 (en) 2009-06-18 2014-04-22 Hoffmann-La Roche, Inc. Bispecific, tetravalent antigen binding proteins
JO3182B1 (ar) 2009-07-29 2018-03-08 Regeneron Pharma مضادات حيوية بشرية عالية الالفة مع تولد الاوعية البشرية - 2
KR101688522B1 (ko) 2009-12-15 2016-12-21 삼성전자주식회사 안지오포이에틴-2에 특이적으로 결합하는 항체 및 그의 용도
WO2012009705A1 (en) 2010-07-15 2012-01-19 Zyngenia, Inc. Ang-2 binding complexes and uses thereof
EP2673297A2 (en) * 2011-02-11 2013-12-18 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof
WO2012162561A2 (en) 2011-05-24 2012-11-29 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
CN105451767B (zh) 2013-03-15 2019-10-18 泽恩格尼亚股份有限公司 多价和单价多特异性复合物及其用途

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150283261A1 (en) * 2011-04-04 2015-10-08 Wisconsin Alumni Research Foundation Method for selective targeting and entry of bacterial toxins to cells
US9623117B2 (en) * 2011-04-04 2017-04-18 Wisconsin Alumni Research Foundation Method for selective targeting and entry of bacterial toxins to cells
US10526381B2 (en) 2011-05-24 2020-01-07 Zygenia, Inc. Multivalent and monovalent multispecific complexes and their uses
US10150800B2 (en) 2013-03-15 2018-12-11 Zyngenia, Inc. EGFR-binding modular recognition domains
US11920128B2 (en) 2013-09-18 2024-03-05 Kymab Limited Methods, cells and organisms
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US10711280B2 (en) 2013-12-11 2020-07-14 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse ES cell genome
US11820997B2 (en) 2013-12-11 2023-11-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US10208317B2 (en) 2013-12-11 2019-02-19 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse embryonic stem cell genome
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US9884913B2 (en) 2014-01-31 2018-02-06 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US9605070B2 (en) 2014-01-31 2017-03-28 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11053307B2 (en) 2014-02-02 2021-07-06 Medimmune Limited Compounds and methods for treating pain
US20150274818A1 (en) * 2014-02-02 2015-10-01 Medlmmune Limited Compounds and methods for treating pain
US11897949B2 (en) 2014-02-02 2024-02-13 Medimmune Limited Compounds and methods for treating pain
US10457728B2 (en) 2014-02-02 2019-10-29 Medimmune Limited Compounds and methods for treating pain
US9884911B2 (en) * 2014-02-02 2018-02-06 Medimmune Limited Compounds and methods for treating pain
US11820818B2 (en) 2014-05-29 2023-11-21 Macrogenics, Inc. Multi-chain polypeptide-containing tri-specific binding molecules
US11697684B2 (en) 2014-05-29 2023-07-11 Macrogenics, Inc. Tri-specific binding molecules that specifically bind to multiple cancer antigens
EP3954703A3 (en) * 2014-05-29 2022-05-18 MacroGenics, Inc. Tri-specific binding molecules and methods of use thereof
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
TWI659042B (zh) * 2014-07-01 2019-05-11 美商輝瑞股份有限公司 雙特異性異質二聚體雙功能抗體及彼等之用途
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US11071771B2 (en) 2014-10-17 2021-07-27 Kodiak Sciences Inc. Butyrylcholinesterase zwitterionic polymer conjugates
WO2016070014A1 (en) * 2014-10-31 2016-05-06 H. Lee Moffitt Cancer Center And Research Institute, Inc. Tetravalent tlr9 bispecific antibody
EP3212232A4 (en) * 2014-10-31 2019-01-23 H. Lee Moffitt Cancer Center And Research Institute, Inc. ANTIBODY BISPECIFIC ANTI-TLR9
WO2016115171A1 (en) * 2015-01-16 2016-07-21 The Regents Of The University Of California Insulin-like growth factor 2 (igf2) signaling and modulation
WO2016145385A3 (en) * 2015-03-11 2016-11-17 Integrated Biotherapeutics, Inc. Pan-ebola and pan-filovirus protective epitopes, antibodies, and antibody cocktails
US10196445B1 (en) * 2015-03-17 2019-02-05 Bristol-Myers Squibb Company Ipilimumab variant with enhanced ADCC
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11591392B2 (en) 2016-07-14 2023-02-28 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
US10533052B2 (en) 2016-07-14 2020-01-14 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
US10077306B2 (en) * 2016-07-14 2018-09-18 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
US11739163B2 (en) * 2016-09-29 2023-08-29 Aebi Ltd. Therapeutic multi-targeting constructs and uses thereof
JP2020522254A (ja) * 2017-05-31 2020-07-30 エルスター セラピューティクス, インコーポレイテッド 骨髄増殖性白血病(mpl)タンパク質に結合する多特異性分子およびその使用
US20190270806A1 (en) * 2018-03-02 2019-09-05 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
US11859001B2 (en) 2020-08-05 2024-01-02 Synthekine, Inc. IL12RB1-Binding molecules and methods of use
US11873349B1 (en) 2020-08-05 2024-01-16 Synthekine, Inc. Compositions and methods related to IL27 receptor binding
WO2022032025A1 (en) * 2020-08-05 2022-02-10 Synthekine, Inc. Ifngr binding synthetic cytokines and methods of use

Also Published As

Publication number Publication date
AU2017204032B2 (en) 2019-10-03
WO2012162561A2 (en) 2012-11-29
DK2714738T3 (en) 2019-01-28
CN106432506A (zh) 2017-02-22
ES2704038T3 (es) 2019-03-13
NZ618334A (en) 2016-05-27
CA2837169A1 (en) 2012-11-29
US20150210765A1 (en) 2015-07-30
CN103857699B (zh) 2016-08-31
NZ719602A (en) 2020-02-28
US20170298106A1 (en) 2017-10-19
AU2012258637B2 (en) 2017-07-20
EP2714738A2 (en) 2014-04-09
WO2012162561A3 (en) 2013-05-02
EP2714738B1 (en) 2018-10-10
AU2012258637A1 (en) 2013-12-19
AU2017204032A1 (en) 2017-07-06
US10526381B2 (en) 2020-01-07
CA2837169C (en) 2021-11-09
CN103857699A (zh) 2014-06-11

Similar Documents

Publication Publication Date Title
AU2017204032B2 (en) Multivalent and monovalent multispecific complexes and their uses
US10150800B2 (en) EGFR-binding modular recognition domains
US10087222B2 (en) Polynucleotides encoding angiopoietin-2 (ang-2) binding polypeptides
US20140088019A1 (en) Monovalent and Multivalent Multispecific Complexes and Uses Thereof
WO2014028776A1 (en) Monovalent and multivalent multispecific complexes and uses thereof
TW201945390A (zh) 靶向cd33之抗體可變域及其用途
EP3755721A1 (en) Multi-specific binding proteins that bind cd33, nkg2d, and cd16, and methods of use
TW202128759A (zh) 結合nkg2d、cd及flt3之蛋白質
NZ719602B2 (en) Multivalent and monovalent multispecific complexes and their uses
NZ618334B2 (en) Multivalent and monovalent multispecific complexes and their uses
EA043040B1 (ru) Пептид, связывающий ангиопоэтин-2, мультиспецифические комплексы, содержащие указанный пептид и антитела, и их применение

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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