US20240018252A1 - Therapeutic antibodies and their uses - Google Patents

Therapeutic antibodies and their uses Download PDF

Info

Publication number
US20240018252A1
US20240018252A1 US18/456,399 US202318456399A US2024018252A1 US 20240018252 A1 US20240018252 A1 US 20240018252A1 US 202318456399 A US202318456399 A US 202318456399A US 2024018252 A1 US2024018252 A1 US 2024018252A1
Authority
US
United States
Prior art keywords
seq
antibody
lymphoma
sequence shown
cell
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.)
Pending
Application number
US18/456,399
Inventor
Tracy Chia-Chien Kuo
Javier Fernando Chaparro Riggers
Wei Chen
Amy Shaw-Ru CHEN
Edward Derrick Pascua
Thomas John VAN BLARCOM
Leila Marie BOUSTANY
Weihsien HO
Yik Andy Yeung
Pavel Strop
Arvind Rajpal
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.)
Pfizer Inc
Original Assignee
Pfizer 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 Pfizer Inc filed Critical Pfizer Inc
Priority to US18/456,399 priority Critical patent/US20240018252A1/en
Publication of US20240018252A1 publication Critical patent/US20240018252A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68031Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being an auristatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6875Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin
    • A61K47/6879Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin the immunoglobulin having two or more different antigen-binding sites, e.g. bispecific or multispecific immunoglobulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or 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
    • C07K2317/30Immunoglobulins 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing 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
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the .xml file contains a sequence listing entitled “PC72209F_SEQListing_ST26.xml” created on Aug. 23, 2023 and having a size of 669 KB.
  • the sequence listing contained in this .xml file is part of the specification and is incorporated herein by reference in its entirety.
  • the present invention relates to antibodies, e.g., full length antibodies or antigen binding fragments thereof, that specifically bind to BCMA (B-Cell Maturation Antigen) and/or CD3 (Cluster of Differentiation 3).
  • the invention also relates to antibody conjugates (e.g., antibody-drug-conjugates) comprising the BCMA antibodies, compositions comprising the BCMA antibodies, and methods of using the BCMA antibodies and their conjugates for treating conditions associated with cells expressing BCMA (e.g., cancer or autoimmune disease).
  • the invention further relates to heteromultimeric antibodies that specifically bind to CD3 and a tumor cell antigen, (e.g., bispecific antibodies that specifically bind to CD3 and BCMA). Compositions comprising such heteromultimeric antibodies, methods for producing and purifying such heterodimeric antibodies, and their use in diagnostics and therapeutics are also provided.
  • B-cell maturation antigen (BCMA, CD269, or TNFRSF17) is a member of the tumor necrosis factor receptor (TNFR) superfamily.
  • BCMA was identified in a malignant human T cell lymphoma containing a t(4;16) translocation. The gene is selectively expressed in the B-cell lineage with the highest expression in plasma blasts and plasma cells, antibody secreting cells.
  • BCMA binds two ligands, B-cell activation factor (BAFF) (also called B-lymphocyte stimulator (BLyS) and APOL-related leukocyte expressed ligand (TALL-1)) and a proliferation-inducing ligand (APRIL) with affinity of 1 uM and 16 nM, respectively.
  • BAFF B-cell activation factor
  • BALL-1 APOL-related leukocyte expressed ligand
  • APRIL proliferation-inducing ligand
  • Binding of APRIL or BAFF to BCMA promotes a signaling cascade involving NF-kappa B, Elk-1, c-Jun N-terminal kinase and the p38 mitogen-activated protein kinase, which produce signals for cell survival and proliferation.
  • BCMA is also expressed on malignant B cells and several cancers that involve B lymphocytes including multiple myeloma, plasmacytoma, Hodgkin's Lymphoma, and chronic lymphocytic leukemia.
  • B lymphocytes including multiple myeloma, plasmacytoma, Hodgkin's Lymphoma, and chronic lymphocytic leukemia.
  • autoimmune diseases where plasmablasts are involved such as systemic lupus erythematosus (SLE) and rheumatoid arthritis
  • BCMA expressing antibody-producing cells secrete autoantibodies that attack self.
  • an alternative treatment to multiple myeloma such as using an anti-BCMA antagonist including antibodies and other immunotherapeutic agents (e.g. bispecific antibodies or antibody-drug conjugates), would make a superior therapeutic agent.
  • an anti-BCMA antagonist including antibodies and other immunotherapeutic agents (e.g. bispecific antibodies or antibody-drug conjugates)
  • the invention disclosed herein is directed to therapeutic antibodies that bind to BCMA and/or CD3.
  • Antibody conjugates e.g., antibody-drug conjugates
  • the heteromultimeric antibodies e.g., bispecific antibodies
  • a tumor cell antigen e.g., bispecific antibodies that specifically bind to CD3 and BCMA
  • the invention provides an isolated antibody, or an antigen binding fragment thereof, which specifically binds to BCMA, wherein the antibody comprises: a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 125, 127, 313, 314, 363, or 365; and/or a VL region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38
  • the VH region comprises (i) a VH CDR1 comprising SEQ ID NO:150, 151, 152, 156, or 157; (ii) a VH CDR2 comprising SEQ ID NO: 169, 154, 194, 159, 195, 196, 162, 158, 198, 177, 178, 199, 200, 201, 202, 203, 204, 206, 207, 208, 172, 203, or 204; and (iii) a VH CDR3 comprising SEQ ID NO: 155, 161, 197, 205, or 164; and/or wherein the VL region comprises (i) a VL CDR1 comprising SEQ ID NO: 209, 271, 273, 275, 251, 277, 260, 279, 245, 283, 285, 287, 290, 292, 235, 297, or 299; (ii) a VL CDR2 comprising SEQ ID NO: 221;
  • the VH region comprises the sequence shown in SEQ ID NO: 112 or a variant with one or several conservative amino acid substitutions in residues that are not within a CDR and/or the VL region comprises the amino acid sequence shown in SEQ ID NO: 38 or a variant thereof with one or several amino acid substitutions in amino acids that are not within a CDR.
  • the antibody comprises a light chain comprising the sequence shown in SEQ ID NO: 357 and a heavy chain comprising the sequence shown in SEQ ID NO: 358.
  • the antibody comprises a VH region produced by the expression vector with ATCC Accession No. PTA-122094.
  • the antibody comprises a VL region produced by the expression vector with ATCC Accession No. PTA-122093.
  • the invention provides an isolated antibody comprising an acyl donor glutamine-containing tag engineered at a specific site of the BCMA antibody of the present invention.
  • the tag comprises an amino acid sequence selected from the group consisting of Q, LQG, LLQGG (SEQ ID NO:318), LLQG (SEQ ID NO:454), LSLSQG (SEQ ID NO: 455), GGGLLQGG (SEQ ID NO: 456), GLLQG (SEQ ID NO: 457), LLQ, GSPLAQSHGG (SEQ ID NO: 458), GLLQGGG (SEQ ID NO: 459), GLLQGG (SEQ ID NO: 460), GLLQ (SEQ ID NO: 461), LLQLLQGA (SEQ ID NO: 462), LLQGA (SEQ ID NO: 463), LLQYQGA (SEQ ID NO: 464), LLQGSG (SEQ ID NO: 465), LLQYQG (SEQ ID NO: 465),
  • the invention provides an isolated antibody comprising an acyl donor glutamine-containing tag and an amino acid modification at position 222, 340, or 370 of the BCMA antibody of the present invention.
  • the amino acid modification is a substitution from lysine to arginine.
  • the BCMA antibody of the present invention further comprises a linker.
  • the linker is selected from the group consisting of Ac-Lys-Gly (acetyl-lysine-glycine), aminocaproic acid, Ac-Lys-p-Ala (acetyl-lysine- ⁇ -alanine), amino-PEG2 (polyethylene glycol)-C2, amino-PEG3-C2, amino-PEG6-C2, Ac-Lys-Val-Cit-PABC (acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl), amino-PEG6-C2-Val-Cit-PABC, aminocaproyl-Val-Cit-PABC, [(3R,5R)-1- ⁇ 3-[2-(2-aminoethoxy)ethoxy]propanoyl ⁇ piperidine-3,5-diyl]bis-Val-Cit-PABC, [(3S,
  • the invention provides a conjugate of the BCMA antibody or the antigen binding fragment as described herein, wherein the antibody or the antigen binding fragment is conjugated to an agent, wherein the agent is selected from the group consisting of a cytotoxic agent, an immunomodulating agent, an imaging agent, a therapeutic protein, a biopolymer, and an oligonucleotide.
  • the agent is a cytotoxic agent including, but not limited to, an anthracycline, an auristatin, a camptothecin, a combretastatin, a dolastatin, a duocarmycin, an enediyne, a geldanamycin, an indolino-benzodiazepine dimer, a maytansine, a puromycin, a pyrrolobenzodiazepine dimer, a taxane, a vinca alkaloid, a tubulysin, a hemiasterlin, a spliceostatin, a pladienolide, and stereoisomers, isosteres, analogs, or derivatives thereof.
  • a cytotoxic agent including, but not limited to, an anthracycline, an auristatin, a camptothecin, a combretastatin, a dolastatin, a duocarmycin, an ened
  • the cytotoxic agent is MMAD (Monomethyl Auristatin D), 0101 (2-methylalanyl-N-[(3R,4S,5S)-3-methoxy-1- ⁇ (2S)-2-[(1R,2R)-1-methoxy-2-methyl-3-oxo-3- ⁇ [(1 S)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino ⁇ propyl]pyrrolidin-1-yl ⁇ -5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), 3377 (N,2-dimethylalanyl-N- ⁇ (1 S,2R)-4- ⁇ (2S)-2-[(1R,2R)-3- ⁇ [(1 S)-1-carboxyl-2-phenylethyl]amino ⁇ -1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl ⁇ -2-methoxy-1-meth
  • the present invention provides a conjugate comprising the formula: antibody-(acyl donor glutamine-containing tag)-(linker)-(cytotoxic agent).
  • the acyl donor glutamine-containing tag comprises an amino acid sequence LLQG (SEQ ID NO: 319) and/or GGLLQGPP (SEQ ID NO: 339) and wherein the linker comprises acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl or amino-PEG6-C2.
  • the conjugate is selected from the group consisting of 1) antibody-GGLLQGPP (SEQ ID NO: 339)-(acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl (AcLys-VC-PABC))-0101; 2) antibody-LLQG (SEQ ID NO: 319)-amino-PEG6-C2-0131; and 3) antibody-LLQG (SEQ ID NO: 319)-amino-PEG6-C2-3377.
  • the conjugate further comprises an amino acid substitution from lysine to arginine at antibody position 222.
  • the conjugate further comprises amino acid substitutions at antibody position N297Q or N297A.
  • a method of producing the BCMA antibody as described herein comprising culturing the host cell under conditions that result in production of the BCMA antibody, and isolating the BCMA antibody from the host cell or culture.
  • the invention provides a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression.
  • a condition e.g., cancer or autoimmune disorder
  • provided is a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting tumor growth or progression.
  • provided is a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting metastasis of malignant cells expressing BCMA.
  • provided is a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inducing tumor regression.
  • the invention provides an isolated antibody, or an antigen binding fragment thereof, which specifically binds to CD3, wherein the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443,445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399.
  • VL light chain variable
  • the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387.
  • VL light chain variable
  • the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 340, 343,
  • the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 407, or 408 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336, 404, 405, or 417; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335 or 406; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343 or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341 or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342 or 439.
  • the antibody comprises a VH region produced by the expression vector with ATCC Accession No. PTA-122513.
  • an isolated antibody which specifically binds to CD3 and competes with the anti-CD3 antibody of the present invention as described herein.
  • the invention provides a bispecific antibody wherein the bispecific antibody is a full-length human antibody, comprising a first antibody variable domain of the bispecific antibody capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen located on the human immune effector cell, and comprising a second antibody variable domain of the bispecific antibody capable of specifically binding to a target antigen, wherein the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in S
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH CDR1-comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 331,
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387; and the second antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 112; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 38.
  • VH heavy chain variable
  • VL light chain variable
  • the second antibody variable domain comprises (a) a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence SYX 1 MX 2 , wherein X 1 is A or P; and X 2 is T, N, or S (SEQ ID NO: 301), GFTFX 1 SY, wherein X 1 is G or S (SEQ ID NO: 302), or GFTFX 1 SYX2MX 3 , wherein X 1 is G or S, X 2 is A or P; and X 3 is T, N, or S (SEQ ID NO: 303); (ii) a VH CDR2 comprising the sequence AX 1 X 2 X 3 X 4 GX 5 X 6 X 7 X 8 YADX 9 X 10 KG, wherein X 1 is I, V, T, H, L, A, or C; X 2 is S, D, G, T, I, L, F,
  • the second antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO:150, 151, 152, 156, 157, 348, 349, 353, 354, or 355; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 169, 154, 194, 159, 195, 196, 162, 158, 198, 177, 178, 199, 200, 201, 202, 203, 204, 206, 207, 208, 172, 203, 204, 350, 351, 356 or 357; and (iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 155, 161, 197, 205, 164, or 352, or 358; and/or wherein the light chain variable (VL) region comprises (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209, 27
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 407, or 408(ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336, 417, 404, or 405; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335 or 406; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343 or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341 or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342 or 439; and (b) the second antibody variable domain comprises a heavy chain VH region comprising (i) a VH
  • both the first and the second antibody variable domains of the bispecific antibody comprise amino acid modifications at positions 223, 225, and 228 in the hinge region and at position 409 or 368 (EU numbering scheme) in the CH3 region of a human IgG2 (SEQ ID NO: 493).
  • the bispecific antibody as described herein further comprises an amino acid modification at position 265 of the human IgG2.
  • the invention provides pharmaceutical compositions comprising any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • antibodies e.g., BCMA, CD3, or bispecific
  • conjugates thereof e.g., BCMA antibody-drug conjugate
  • the invention also provides cell lines that recombinantly produce any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • antibodies e.g., BCMA, CD3, or bispecific
  • conjugates thereof e.g., BCMA antibody-drug conjugate
  • the invention also provides nucleic acids encoding any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • the invention also provides nucleic acids encoding a heavy chain variable region and/or a light chain variable region of any of these antibodies described herein.
  • kits comprising an effective amount of any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • antibodies e.g., BCMA, CD3, or bispecific
  • conjugates thereof e.g., BCMA antibody-drug conjugate
  • the invention also provides methods of treating a condition (e.g., tumor growth/progression inhibition; metastasis of malignant cells expressing BCMA inhibition; tumor regression induction in subjects with malignant cells expressing BCMA) in subjects in need thereof comprising providing the isolated antibodies (e.g., BCMA) or binding fragments, bispecific antibodies (BCMA-CD3 bispecifics), or the conjugates thereof (e.g., BCMA antibody-drug conjugates) thereof described herein and administering said antibodies or conjugates to said subject.
  • a condition e.g., tumor growth/progression inhibition; metastasis of malignant cells expressing BCMA inhibition; tumor regression induction in subjects with malignant cells expressing BCMA
  • the condition is cancer.
  • the cancer is a B-cell related cancer selecting from the group consisting of multiple myeloma, malignant plasma cell neoplasm, Hodgkin's lymphoma, nodular lymphocyte predominant Hodgkin's lymphoma, Kahler's disease and Myelomatosis, plasma cell leukemia, plasmacytoma, B-cell prolymphocytic leukemia, hairy cell leukemia, B-cell non-Hodgkin's lymphoma (NHL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), follicular lymphoma, Burkitt's lymphoma
  • the antibodies described herein comprise a constant region. In some embodiments, the antibodies described herein are of the human IgG1, IgG2 or IgG2Aa, IgG3, or IgG4 subclass. In some embodiments, the antibodies described herein comprise a glycosylated constant region. In some embodiments, the antibodies described herein comprise a constant region having increased binding affinity to one or more human Fc gamma receptor(s).
  • FIG. 1 A - FIG. 1 D depict the double-referenced sensorgrams with fit curves for interactions between selected anti-BCMA antibodies of the present invention and human BCMA.
  • FIG. 2 depicts in vivo efficacy studies of various anti-BCMA ADCs in the MM1 S orthotopic multiple myeloma model, including P6E01_VHVL-AcLys-Val-Cit-PABC-Aur0101; P5A2_VHVL-AcLys-Val-Cit-PABC-Aur0101; P5C1_VHVL-AcLys-Val-Cit-PABC-Aur0101; P4G4-AcLys-Val-Cit-PABC-Aur0101; and P1A11-AcLys-Val-Cit-PABC-Aur0101.
  • NNC is a negative control non-BCMA antibody.
  • “LCQ05” and “LCQ04” correspond to glutamine-containing transglutaminase tag SEQ ID NOs: 474 and 475, respectively.
  • FIG. 3 depicts in vivo efficacy of the anti-BCMA ADCs in the MM1S orthotopic multiple myeloma model, including L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101.
  • NNC is a control non-BCMA antibody.
  • “LCQ05” and H7c correspond to glutamine-containing transglutaminase tag SEQ ID NO: 474 and SEQ ID NO: 454, respectively
  • FIG. 4 also depicts in vivo efficacy of the anti-BCMA ADCs in the MM1S orthotopic multiple myeloma model, including L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-Aur0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101.
  • NNC is a control non-BCMA antibody (antibody-N297Q/K222R-AcLys-VC-PABC-0101).
  • LCQ05 and H7c correspond to glutamine-containing transglutaminase tag SEQ ID NO: 474 and SEQ ID NO: 454, respectively.
  • FIG. 5 also depicts in vivo efficacy of an anti-BCMA ADC in the MM1S orthotopic multiple myeloma model.
  • Anti-BCMA antibody COMBO_Rd4_0.6 nM-C29 (“Combo C29 DI) is conjugated to H7c/N297A/K222R-amino-PEG6-C2-131 at doses ranging from 0.1 mg/kg, 0.38 mg/kg, 0.75 mg/kg, 1.5 mg/kg in comparison to NNC, a control non-BCMA antibody (antibody-N297Q/K222R-AcLys-VC-PABC-0101) at 3 mg/kg.
  • H7c correspond to glutamine-containing transglutaminase tag SEQ ID NO: 454.
  • FIG. 6 A - FIG. 6 F depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. B cell depletion following a single dose of bispecific antibody is shown as a percentage of prestudy counts.
  • FIG. 7 A - FIG. 7 F depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. CD8+ T cell kinetics were tracked following a single dose of bispecific antibody.
  • FIG. 8 A and FIG. 8 B depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys.
  • the effect of the monovalent CD3 antibody on T cell kinetics and proliferation was analyzed.
  • FIGS. 9 A - FIG. 9 D depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. The effect of anti-CD3 arm affinity on B cell depletion was analyzed.
  • FIGS. 10 A and 10 B show that the selected anti-CD3 antibodies had Thymidine incorporation reading on human and cynomolgus PBMC.
  • FIG. 11 A - FIG. 11 D show that all human anti-EpCam_h2B4 bispecific antibodies have cell killing activity on in vitro setting.
  • FIG. 12 shows that a single dose of human anti-BCMA/CD3 bispecific antibody resulted in tumor regression in a dose-dependent manner in an orthotopic MM1.S myeloma model.
  • FIG. 13 shows that two doses of human anti-BCMA/CD3 bispecific antibody resulted in increased tumor regression in an orthotopic Molp8 myeloma model.
  • FIG. 14 shows that anti-BCMA/CD3 bispecific antibody alone or in combination with standard of care for multiple myeloma (lenalidomide or bortezomib) is more efficacious than lenalidomide and bortezomib combined in orthotopic Molp8 tumor model.
  • FIG. 15 A - FIG. 15 C show that carfilzomib, lenalidomide, and doxorubicin do not have a negative effect on the function of the anti-BCMA/CD3 bispecific antibody on OPM2 cells as compared to the anti-BCMA/CD3 bispecific antibody alone.
  • FIG. 16 shows synergistic effects on the function of anti-BCMA/CD3 bispecific antibody when combined with carfilzomib and lenalidomide in comparison to each molecule alone.
  • the invention disclosed herein provides antibodies and antibody conjugates (e.g., antibody-drug conjugates) that specifically bind to BCMA (e.g., human BCMA).
  • the invention also provides polynucleotides encoding these antibodies and conjugates, compositions comprising these antibodies and conjugates, and methods of making these antibodies and conjugates.
  • the invention disclosed herein provides antibodies that specifically bind to CD3 (e.g., human CD3) as well as heterodimeric antibodies (e.g., bispecific antibodies) that specifically bind to CD3 and a tumor antigen (e.g., BCMA).
  • the invention also provides polynucleotides encoding these antibodies, compositions comprising these antibodies, and methods of making and using these antibodies.
  • the invention further provides methods for treating a condition associated with malignant BCMA expression in a subject, such as cancer or autoimmune disease, using the antibodies (e.g., BCMA, CD3, or bispecific antibody) or conjugates thereof (BCMA antibody-drug conjugates) as described herein.
  • a condition associated with malignant BCMA expression in a subject such as cancer or autoimmune disease
  • the antibodies e.g., BCMA, CD3, or bispecific antibody
  • conjugates thereof BCMA antibody-drug conjugates
  • an “antibody” is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • a target such as a carbohydrate, polynucleotide, lipid, polypeptide, etc.
  • the term encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab′, F(ab′)2, Fv), single chain (ScFv) and domain antibodies (including, for example, shark and camelid antibodies), and fusion proteins comprising an antibody, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site.
  • An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • the heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • antigen binding fragment or “antigen binding portion” of an antibody, as used herein, refers to one or more fragments of an intact antibody that retain the ability to specifically bind to a given antigen (e.g., BCMA or CD3). Antigen binding functions of an antibody can be performed by fragments of an intact antibody.
  • binding fragments encompassed within the term “antigen binding fragment” of an antibody include Fab; Fab′; F(ab′)2; an Fd fragment consisting of the VH and CH1 domains; an Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a single domain antibody (dAb) fragment (Ward et al., Nature 341:544-546, 1989), and an isolated complementarity determining region (CDR).
  • An antibody, an antibody conjugate, or a polypeptide that “preferentially binds” or “specifically binds” (used interchangeably herein) to a target is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art.
  • a molecule is said to exhibit “specific binding” or “preferential binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances.
  • an antibody that specifically or preferentially binds to a BCMA epitope or CD3 epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other BCMA epitopes, non-BCMA epitopes, CD3 epitopes, or non-CD3 epitopes.
  • an antibody or moiety or epitope that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target.
  • “specific binding” or “preferential binding” does not necessarily require (although it can include) exclusive binding.
  • reference to binding means preferential binding.
  • variable region of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions.
  • FR framework regions
  • CDRs complementarity determining regions
  • the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies.
  • There are at least two techniques for determining CDRs (1) an approach based on cross-species sequence variability (i.e., Kabat et al.
  • a CDR may refer to CDRs defined by either approach or by a combination of both approaches.
  • a “CDR” of a variable domain are amino acid residues within the variable region that are identified in accordance with the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art.
  • Antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See, e.g., Kabat et al., 1992, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH, Washington D.C. The positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others.
  • CDR identification includes the “AbM definition,” which is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys®), or the “contact definition” of CDRs based on observed antigen contacts, set forth in MacCallum et al., J. Mol. Biol., 262:732-745, 1996.
  • the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding.
  • a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
  • “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, Nature 256:495, 1975, or may be made by recombinant DNA methods such as described in U.S. Pat. No. 4,816,567.
  • the monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et al., Nature 348:552-554, 1990, for example.
  • humanized antibody refers to forms of non-human (e.g. murine) antibodies that are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab′, F(ab′)2 or other antigen binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementarity determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • CDR complementarity determining region
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • CDR L1, CDR L2, CDR L3, CDR H1, CDR H2, or CDR H3 are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody.
  • human antibody means an antibody having an amino acid sequence corresponding to that of an antibody produced by a human and/or which has been made using any of the techniques for making human antibodies known to those skilled in the art or disclosed herein.
  • This definition of a human antibody includes antibodies comprising at least one human heavy chain polypeptide or at least one human light chain polypeptide.
  • One such example is an antibody comprising murine light chain and human heavy chain polypeptides.
  • Human antibodies can be produced using various techniques known in the art. In one embodiment, the human antibody is selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., Nature Biotechnology, 14:309-314, 1996; Sheets et al., Proc. Natl. Acad.
  • Human antibodies can also be made by immunization of animals into which human immunoglobulin loci have been transgenically introduced in place of the endogenous loci, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. This approach is described in U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016.
  • the human antibody may be prepared by immortalizing human B lymphocytes that produce an antibody directed against a target antigen (such B lymphocytes may be recovered from an individual or from single cell cloning of the cDNA, or may have been immunized in vitro). See, e.g., Cole et al. Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77, 1985; Boerner et al., J. Immunol., 147 (1):86-95, 1991; and U.S. Pat. No. 5,750,373.
  • chimeric antibody is intended to refer to antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody.
  • polypeptide oligopeptide
  • peptide protein
  • the terms “polypeptide”, “oligopeptide”, “peptide” and “protein” are used interchangeably herein to refer to chains of amino acids of any length, preferably, relatively short (e.g., 10-100 amino acids).
  • the chain may be linear or branched, it may comprise modified amino acids, and/or may be interrupted by non-amino acids.
  • the terms also encompass an amino acid chain that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • polypeptides can occur as single chains or associated chains.
  • a “monovalent antibody” comprises one antigen binding site per molecule (e.g., IgG or Fab). In some instances, a monovalent antibody can have more than one antigen binding sites, but the binding sites are from different antigens.
  • a “monospecific antibody” comprises two identical antigen binding sites per molecule (e.g. IgG) such that the two binding sites bind identical epitope on the antigen. Thus, they compete with each other on binding to one antigen molecule. Most antibodies found in nature are monospecific. In some instances, a monospecific antibody can also be a monovalent antibody (e.g. Fab)
  • a “bivalent antibody” comprises two antigen binding sites per molecule (e.g., IgG).
  • the two binding sites have the same antigen specificities.
  • bivalent antibodies may be bispecific.
  • bispecific or dual-specific is a hybrid antibody having two different antigen binding sites.
  • the two antigen binding sites of a bispecific antibody bind to two different epitopes, which may reside on the same or different protein targets.
  • a “bifunctional” is antibody is an antibody having identical antigen binding sites (i.e., identical amino acid sequences) in the two arms but each binding site can recognize two different antigens.
  • heteromultimer is a molecule comprising at least a first polypeptide and a second polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue.
  • the heteromultimer can comprise a “heterodimer” formed by the first and second polypeptide or can form higher order tertiary structures where polypeptides in addition to the first and second polypeptide are present.
  • heterodimer is a molecule comprising a first polypeptide and a second polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue.
  • the “hinge region,” “hinge sequence”, and variations thereof, as used herein, includes the meaning known in the art, which is illustrated in, for example, Janeway et al., ImmunoBiology: the immune system in health and disease, (Elsevier Science Ltd., NY) (4th ed., 1999); Bloom et al., Protein Science (1997), 6:407-415; Humphreys et al., J. Immunol. Methods (1997), 209:193-202.
  • immunoglobulin-like hinge region refers to the hinge region and hinge sequence of an immunoglobulin-like or an antibody-like molecule (e.g., immunoadhesins).
  • the immunoglobulin-like hinge region can be from or derived from any IgG1, IgG2, IgG3, or IgG4 subtype, or from IgA, IgE, IgD or IgM, including chimeric forms thereof, e.g., a chimeric IgG1/2 hinge region.
  • immune effector cell refers to a cell within the natural repertoire of cells in the human immune system which can be activated to affect the viability of a target cell.
  • the viability of a target cell can include cell survival, proliferation, and/or ability to interact with other cells.
  • Antibodies of the invention can be produced using techniques well known in the art, e.g., recombinant technologies, phage display technologies, synthetic technologies or combinations of such technologies or other technologies readily known in the art (see, for example, Jayasena, S. D., Clin. Chem., 45: 1628-50, 1999 and Fellouse, F. A., et al, J. Mol. Biol., 373(4):924-40, 2007).
  • polynucleotide or “nucleic acid,” as used interchangeably herein, refer to chains of nucleotides of any length, and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a chain by DNA or RNA polymerase.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the chain.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • Other types of modifications include, for example, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metal
  • any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports.
  • the 5′ and 3′ terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms.
  • Other hydroxyls may also be derivatized to standard protecting groups.
  • Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2′-O-methyl-, 2′-O-allyl, 2′-fluoro- or 2′-azido-ribose, carbocyclic sugar analogs, alpha- or beta-anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and abasic nucleoside analogs such as methyl riboside.
  • One or more phosphodiester linkages may be replaced by alternative linking groups.
  • linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S(“thioate”), P(S)S (“dithioate”), (O)NR 2 (“amidate”), P(O)R, P(O)OR′, CO or CH 2 (“formacetal”), in which each R or R′ is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (—O—) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • a “constant region” of an antibody refers to the constant region of the antibody light chain or the constant region of the antibody heavy chain, either alone or in combination.
  • substantially pure refers to material which is at least 50% pure (i.e., free from contaminants), more preferably, at least 90% pure, more preferably, at least 95% pure, yet more preferably, at least 98% pure, and most preferably, at least 99% pure.
  • a “host cell” includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts.
  • Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation.
  • a host cell includes cells transfected in vivo with a polynucleotide(s) of this invention.
  • the term “Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain.
  • the “Fc region” may be a native sequence Fc region or a variant Fc region.
  • the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof.
  • the numbering of the residues in the Fc region is that of the EU index as in Kabat. Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991.
  • the Fc region of an immunoglobulin generally comprises two constant regions, CH2 and CH3.
  • Fc receptor and “FcR” describe a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA (an “activating receptor”) and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • FcRs are reviewed in Ravetch and Kinet, Ann. Rev. Immunol., 9:457-92, 1991; Capel et al., Immunomethods, 4:25-34, 1994; and de Haas et al., J. Lab. Clin. Med., 126:330-41, 1995.
  • FcR also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol., 117:587, 1976; and Kim et al., J. Immunol., 24:249, 1994).
  • the term “compete”, as used herein with regard to an antibody means that a first antibody, or an antigen binding fragment (or portion) thereof, binds to an epitope in a manner sufficiently similar to the binding of a second antibody, or an antigen binding portion thereof, such that the result of binding of the first antibody with its cognate epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody.
  • the alternative, where the binding of the second antibody to its epitope is also detectably decreased in the presence of the first antibody can, but need not be the case. That is, a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope.
  • each antibody detectably inhibits the binding of the other antibody with its cognate epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to “cross-compete” with each other for binding of their respective epitope(s).
  • Both competing and cross-competing antibodies are encompassed by the present invention. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate, based upon the teachings provided herein, that such competing and/or cross-competing antibodies are encompassed and can be useful for the methods disclosed herein.
  • a “functional Fc region” possesses at least one effector function of a native sequence Fc region.
  • effector functions include Clq binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity; phagocytosis; down-regulation of cell surface receptors (e.g. B cell receptor), etc.
  • Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
  • a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • a “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region.
  • the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably, from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
  • the variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably, at least about 90% sequence identity therewith, more preferably, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% sequence identity therewith.
  • effector function refers to the biological activities attributable to the Fc region of an antibody.
  • antibody effector functions include, but are not limited to, antibody-dependent cell-mediated cytotoxicity (ADCC), Fc receptor binding, complement dependent cytotoxicity (CDC), phagocytosis, Clq binding, and down regulation of cell surface receptors (e.g., B cell receptor; BCR). See, e.g., U.S. Pat. No. 6,737,056.
  • Such effector functions generally require the Fc region to be combined with a binding domain (e.g., an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
  • An exemplary measurement of effector function is through Fc ⁇ 3 and/or Clq binding.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs Fc receptors
  • NK natural killer cells
  • macrophages e.g. natural killer cells, neutrophils, and macrophages
  • ADCC activity of a molecule of interest can be assessed using an in vitro ADCC assay, such as that described in U.S. Pat. No. 5,500,362 or 5,821,337.
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and NK cells.
  • PBMC peripheral blood mononuclear cells
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes et al., 1998, PNAS (USA), 95:652-656.
  • “Complement dependent cytotoxicity” or “CDC” refers to the lysing of a target in the presence of complement.
  • the complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule (e.g. an antibody) complexed with a cognate antigen.
  • a CDC assay e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods, 202: 163 (1996), may be performed.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cells, inhibiting metastasis of neoplastic cells, remission of a BCMA associated disease (e.g., cancer or autoimmune disease), decreasing symptoms resulting from a BCMA associated disease (e.g., cancer or autoimmune disease), increasing the quality of life of those suffering from a BCMA associated disease (e.g., cancer or autoimmune disease), decreasing the dose of other medications required to treat a BCMA associated disease (e.g., cancer or autoimmune disease), delaying the progression of a BCMA associated disease (e.g., cancer or autoimmune disease), curing a BCMA associated disease (e.g., cancer or autoimmune disease), and/or prolong survival of patients having a BCMA associated disease (e.g., cancer or autoimmune disease). “Ameliorating”
  • an “effective dosage” or “effective amount” of drug, compound, or pharmaceutical composition is an amount sufficient to effect any one or more beneficial or desired results.
  • beneficial or desired results include eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results include clinical results such as reducing incidence or amelioration of one or more symptoms of various BCMA associated diseases or conditions (such as multiple myeloma), decreasing the dose of other medications required to treat the disease, enhancing the effect of another medication, and/or delaying the progression of the BCMA associated disease of patients.
  • An effective dosage can be administered in one or more administrations.
  • an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
  • an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • mammals are a mammal, more preferably, a human. Mammals also include, but are not limited to, farm animals, sport animals, pets, primates, horses, dogs, cats, mice and rats.
  • vector means a construct, which is capable of delivering, and, preferably, expressing, one or more gene(s) or sequence(s) of interest in a host cell.
  • vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells.
  • expression control sequence means a nucleic acid sequence that directs transcription of a nucleic acid.
  • An expression control sequence can be a promoter, such as a constitutive or an inducible promoter, or an enhancer.
  • the expression control sequence is operably linked to the nucleic acid sequence to be transcribed.
  • “pharmaceutically acceptable carrier” or “pharmaceutical acceptable excipient” includes any material which, when combined with an active ingredient, allows the ingredient to retain biological activity and is non-reactive with the subject's immune system. Examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents. Preferred diluents for aerosol or parenteral administration are phosphate buffered saline (PBS) or normal (0.9%) saline. Compositions comprising such carriers are formulated by well known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, P A, 1990; and Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005).
  • PBS phosphate buffered saline
  • Compositions comprising such carriers are formulated by well
  • acyl donor glutamine-containing tag or “glutamine tag” as used herein refers to a polypeptide or a protein containing one or more Gln residue(s) that acts as a transglutaminase amine acceptor. See, e.g., WO2012059882 and WO2015015448.
  • k on or “k a ”, as used herein, refers to the rate constant for association of an antibody to an antigen. Specifically, the rate constants (k on /k a and k off /k d ) and equilibrium dissociation constants are measured using whole antibody (i.e. bivalent) and monomeric BCMA proteins.
  • k on or “k d ”, as used herein, refers to the rate constant for dissociation of an antibody from the antibody/antigen complex.
  • K D refers to the equilibrium dissociation constant of an antibody-antigen interaction.
  • references to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X.” Numeric ranges are inclusive of the numbers defining the range.
  • the present invention encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group, but 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 claimed invention.
  • the present invention provides an antibody that binds to BCMA (e.g., human BCMA (e.g., SEQ ID NO: 353 or accession number: Q02223-2) and characterized by any one or more of the following characteristics: (a) treat, prevent, ameliorate one or more symptoms of a condition associated with malignant cells expressing BCMA in a subject (e.g., B-cell related cancer such as multiple myeloma); (b) inhibit tumor growth or progression in a subject (who has a malignant tumor expressing BCMA); (c) inhibit metastasis of cancer (malignant) cells expressing BCMA in a subject (who has one or more malignant cells expressing BCMA); (f) induce regression (e.g., long-term regression) of a tumor expressing BCMA; (d) exert cytotoxic activity in malignant cells expressing BCMA; and (e) block BCMA interaction with other yet to be identified factors.
  • BCMA e.g., human BCMA (e.g., SEQ ID NO: 3
  • an isolated antibody, or an antigen binding fragment thereof, which specifically binds to BCMA wherein the antibody comprises: a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 112, 125, 127, 313, 314, 363, or 365; and/or a VL region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36,
  • an antibody having any one of partial light chain sequence as listed in Table 1 and/or any one of partial heavy chain sequence as listed in Table 1.
  • underlined sequences are CDR sequences according to Kabat and in bold according to Chothia, except for the following heavy chain CDR2 sequences, in which the Chothia CDR sequences are underlined and the Kabat CDR sequences are in bold:
  • the invention also provides CDR portions of antibodies to BCMA (including Chothia, Kabat CDRs, and CDR contact regions). Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed “combined CRs” or “extended CDRs”). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof. Table 2 provides examples of CDR sequences provided herein.
  • H3.AQ SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIAAQMDY For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 135) following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133) mAbs: NO: 130) (Chothia); (Chothia) P6E01/H3.AQ; GFTFGSYAMT (SEQ L1.LGF/L3. ID NO: 131) KW/H3.AQ; (extended) L1.LGF/L3. PY/H3.AQ; L1.LGF/L3. NY/H3.AQ; L1.GDF/L3. KW/H3.AQ; L1.GDF/L3.
  • H2.QR SYAMT SEQ ID NO: AISGSGGNTFYADQRKG VSPIASGMDY For the 129) (Kabat); (SEQ ID NO: 138) (Kabat) (SEQ ID NO: 134) following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133) mAbs: NO: 130) (Chothia); (Chothia) L3.PY/H2. GFTFGSYAMT (SEQ QR; ID NO: 131) L3.PY/L1. (extended) PS/H2.QR; L3.PY/L1. AH/H2.QR; L3.PY/L1. FF/H2.QR; L3.PY/L1.
  • H2.YQ SYAMT SEQ ID NO: AISYQGGNTFYADSVKG VSPIASGMDY For the 129) (Kabat); (SEQ ID NO: 141) (Kabat) (SEQ ID NO: 134) following GFTFGSY (SEQ ID SYQGGN (SEQ ID NO: 142) mAbs: NO: 130) (Chothia); (Chothia) L3.PY/H2.YQ; GFTFGSYAMT (SEQ L3.PY/L1. ID NO: 131) PS/H2.YQ; (extended) L3.PY/L1.
  • PS/H2.LT (extended) L3.PY/L1. AH/H2.LT; L3.PY/L1. FF/H2.LT; L3.PY/L3. KY/H2.LT; and L3.PY/L3. KF/H2.LT.
  • H2.HA SYAMT SEQ ID NO: AISHAGGNTFYADSVKG VSPIASGMDY For the 129) (Kabat); (SEQ ID NO: 145) (Kabat) (SEQ ID NO: 134) following GFTFGSY (SEQ ID SHAGGN (SEQ ID NO: 146) mAbs: NO: 130) (Chothia); (Chothia) L3.PY/H2.HA; GFTFGSYAMT (SEQ L3.PY/L1. ID NO: 131) AH/H2.HA; (extended) L3.PY/L1. FF/H2.HA; L3.PY/L1. PH/H2.HA; and L3.PY/L3. KY/H2.HA.
  • H2.QL SYAMT (SEQ ID NO: AISGSGGNTFYADQLKG VSPIASGMDY For the 129) (Kabat); (SEQ ID NO: 147) (Kabat) (SEQ ID NO: 134) following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133) mAbs: NO: 130) (Chothia); (Chothia) L3.PY/H2.QL; GFTFGSYAMT (SEQ L3.PY/L1. ID NO: 131) PS/H2.QL; (extended) L3.PY/L1. AH/H2.QL; L3.PY/L1. FF/H2.QL; L3.PY/L3.
  • P5A2_VH SYAMN (SEQ ID NO: AISDSGGSTYYADSVKG YWPMDI (SEQ ID VL and 150) (Kabat); (SEQ ID NO: 153) (Kabat) NO: 155) A02_Rd4_ GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154) 6nM_C03 NO: 151) (Chothia); (Chothia) GFTFSSYAMN (SEQ ID NO: 152) (extended) COMBO SYPMS (SEQ ID NO: AIGGSGGSLPYADSVKG YWPMDI (SEQ ID Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 158) (Kabat) NO: 155) C17;
  • L1.LGF/ RASQSLGSFYLA GASSRAT SEQ ID NO: KHYGWPPSFT L3.KW (SEQ ID NO: 212) 210) (SEQ ID NO: 213)
  • L1.LGF/ L3.KW/P6E01 L1.LGF/ L3.KW/H3.AL
  • L1.GDF/ L3.NY/H3.AQ L1.LGF/ RASQSLGSFYLA GASSRAT SEQ ID NO: QHYPYPPSFT L3.PY (SEQ ID NO: 212) 210) (SEQ ID NO: 216)
  • SEQ ID NO: 212 For the following mAbs: L1.LGF/ L3.PY/H3. AP; and L1.LGF/ L3.PY/H3.
  • mAbs L3.PY/L1. AH/P6E01; L3.PY/L1. AH/H2.QR; L3.PY/L1. AH/H2.DY; L3.PY/L1.
  • FF/H2.QR L3.PY/L1. FF/H2.DY; L3.PY/L1. FF/H2.YQ; L3.PY/L1. FF/H2.LT; L3.PY/L1. FF/H2.HA; L3.PY/L1. FF/H2.QL; L3.PY/L1. FF/H3.YA; L3.PY/L1. FF/H3.AE; L3.PY/L1. FF/H3.AQ; and L3.PY/L1.
  • PH/H3.TAQ L3.PY/ RASQSVSSSYLA GASSRAT SEQ ID NO: KYYPYPPSFT L3.KY (SEQ ID NO: 209) 210) (SEQ ID NO: 220)
  • L3.PY/L3. KY/P6E01 L3.PY/L3. KY/H2.QR; L3.PY/L3. KY/H2.DY; L3.PY/L3. KY/H2.YQ; L3.PY/L3. KY/H2.LT; L3.PY/L3. KY/H2.HA; L3.PY/L3. KY/H2.QL; L3.PY/L3. KY/H3.YA; and L3.PY/L3.
  • the present invention provides an antibody that binds to BCMA and competes with the antibody as described herein, including P6E01/P6E01, P6E01/H3.AQ, Li.LGF/L3.KW/P6E01; Li.LGF/L3.NY/P6E01, Li.GDF/L3.NY/P6E01, Li.LGF/L3.KW/H3.AL, Li.LGF/L3.KW/H3.AP, Li.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.KW/
  • the present invention provides an antibody or an antigen binding fragment, which specifically binds to BCMA, wherein the antibody comprises a VH region comprising a sequence shown in SEQ ID NO: 112; and/or a VL region comprising a sequence shown in SEQ ID NO: 38.
  • the antibody comprises a light chain comprising the sequence EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLMYDASIRATG IPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYQSWPLTFGQGTKVEIKRTVAAPSV FIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDST YSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 357) and a heavy chain comprising the sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYPMSWVRQAPGKGLEWVSAIGGSGG SLPYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARYWPMDIWGQGTLVTV SSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTS
  • the present invention provides an antibody or an antigen binding fragment, which specifically bind to BCMA, wherein the antibody comprises a VH region comprising a sequence shown in SEQ ID NO: 2, 32, 42, or 78; and/or a VL region comprising a sequence shown in SEQ ID NO: 6, 16, 43, or 85.
  • the invention also provides CDR portions of antibodies to BCMA antibodies based on CDR contact regions.
  • CDR contact regions are regions of an antibody that imbue specificity to the antibody for an antigen.
  • CDR contact regions include the residue positions in the CDRs and Vernier zones which are constrained in order to maintain proper loop structure for the antibody to bind a specific antigen. See, e.g., Makabe et al., J. Biol. Chem., 283:1156-1166, 2007. Determination of CDR contact regions is well within the skill of the art.
  • the binding affinity (K D ) of the BCMA antibody as described herein to BCMA can be about 0.002 nM to about 6500 nM.
  • the binding affinity is about any of 6500 nm, 6000 nm, 5986 nm, 5567 nm, 5500 nm, 4500 nm, 4000 nm, 3500 nm, 3000 nm, 2500 nm, 2134 nm, 2000 nm, 1500 nm, 1000 nm, 750 nm, 500 nm, 400 nm, 300 nm, 250 nm, 200 nM, 193 nM, 100 nM, 90 nM, 50 nM, 45 nM, 40 nM, 35 nM, 30 nM, 25 nM, 20 nM, 19 nm, 18 nm, 17 nm, 16 nm, 15
  • the binding affinity is less than about any of 6500 nm, 6000 nm, 5500 nm, 5000 nm, 4000 nm, 3000 nm, 2000 nm, 1000 nm, 900 nm, 800 nm, 250 nM, 200 nM, 100 nM, 50 nM, 30 nM, 20 nM, 10 nM, 7.5 nM, 7 nM, 6.5 nM, 6 nM, 5 nM, 4.5 nM, 4 nM, 3.5 nM, 3 nM, 2.5 nM, 2 nM, 1.5 nM, 1 nM, or 0.5 nM.
  • compositions comprising antibodies described herein or made by the methods and having the characteristics described herein.
  • compositions comprise one or more antibodies that bind to BCMA, and/or one or more polynucleotides comprising sequences encoding one or more these antibodies.
  • compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • the invention also provides methods of making any of these antibodies.
  • the antibodies of this invention can be made by procedures known in the art.
  • the polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis.
  • Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available.
  • an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Pat. Nos. 5,807,715; 4,816,567; and 6,331,415.
  • fusion proteins comprising one or more fragments or regions from the antibodies of this invention.
  • a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of the variable light chain region shown in SEQ ID NOs: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38, 40, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 317, 81, 82, 84, 85, 86, 88, 89, 90, 91, 93, 94, 96, 98, 100, 102, 103, 105, 107, 108, 109, 111, 113, 115, 116, 117, 119, 121, 123, 124, 126, 128, 80, 315, 36, or 364, and/or at least 10 amino acids of the variable heavy chain region
  • a fusion polypeptide comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region.
  • the fusion polypeptide comprises a light chain variable region and/or a heavy chain variable region, as shown in any of the sequence pairs selected from among SEQ ID NOs: 1 and 2, 1 and 3, 4 and 2, 5 and 2, 6 and 2, 4 and 7, 4 and 8, 4 and 3, 9 and 8, 9 and 3, 10 and 7, 10 and 8, 10 and 3, 11 and 7, 11 and 8, 11 and 3, 12 and 3, 13 and 7, 13 and 8, 14 and 3, 15 and 2, 16 and 2, 17 and 2, 18 and 2, 19 and 2, 20 and 2, 21 and 2, 22 and 2, 23 and 2, 16 and 24, 16 and 25, 16 and 26, 16 and 27, 16 and 28, 16 and 29, 16 and 30, 16 and 31, 16 and 3, 16 and 32, 16 and 2, 18 and 24, 18 and 25, 18 and 26, 18 and 27, 18 and 28, 18 and 29, 18 and 30, 18 and 31, 18 and 3, 18 and 32, 19 and 24, 19 and 25, 19 and 26, 19 and 27, 19 and 28, 19 and 29, 19 and 30, 19 and 31, 19 and 3, 19 and 32, 20 and 24, 20 and 25, 20 and 26, 20 and 27, 20 and 28, 20 and 29, 20 and 30, 20 and 31, 20 and 3, 20 and 3, 20
  • the fusion polypeptide comprises one or more CDR(s).
  • the fusion polypeptide comprises CDR H3 (VH CDR3) and/or CDR L3 (VL CDR3).
  • a fusion protein contains one or more antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region.
  • heterologous sequences include, but are not limited to a “tag” such as a FLAG tag or a 6His tag. Tags are well known in the art.
  • the invention also provides isolated polynucleotides encoding the antibodies of the invention, and vectors and host cells comprising the polynucleotide.
  • a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody P6E01/P6E01, P6E01/H3.AQ, L1.LGF/L3.KW/P6E01, L1.LGF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.LGF/L3.KW/H3.AL, L1.LGF/L3.KW/H3.AP, L1.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/
  • the invention also encompasses scFv of antibodies of this invention.
  • Single chain variable region fragments are made by linking light and/or heavy chain variable regions by using a short linking peptide (Bird et al., Science 242:423-426, 1988).
  • An example of a linking peptide is (GGGGS) 3 (SEQ ID NO: 498), which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region.
  • Linkers of other sequences have been designed and used (Bird et al., 1988, supra). Linkers should be short, flexible polypeptides and preferably comprised of less than about 20 amino acid residues.
  • Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports.
  • the single chain variants can be produced either recombinantly or synthetically.
  • an automated synthesizer can be used for synthetic production of scFv.
  • a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli .
  • Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides.
  • the resultant scFv can be isolated using standard protein purification techniques known in the art.
  • Diabodies are bivalent, bispecific antibodies in which heavy chain variable (VH) and light chain variable (VL) domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al., Proc. Natl. Acad Sci. USA 90:6444-6448, 1993; Poljak, R. J., et al., Structure 2:1121-1123, 1994).
  • Minibody includes the VL and VH domains of a native antibody fused to the hinge region and CH3 domain of the immunoglobulin molecule. See, e.g., U.S. Pat. No. 5,837,821.
  • the invention provides compositions (such as a pharmaceutical compositions) comprising any of the polynucleotides of the invention.
  • the composition comprises an expression vector comprising a polynucleotide encoding any of the antibodies described herein.
  • the composition comprises either or both of the polynucleotides shown in SEQ ID NO: 486 and SEQ ID NO: 485 below:
  • COMBO_Rd4_0.6nM_C29 heavy chain variable region (SEQ ID NO: 486) GAAGTCCAACTCCTCGAATCCGGTGGCGGCCTTGTCCAGCCTGGAGGTT CCTTGCGCCTGTCATGTGCCGCCAGCGGATTCACCTTCTCGTCCTACCC GATGTCGTGGGTCCGCCAGGCTCCGGGAAAGGGCCTGGAATGGGTGTCA GCCATCGGAGGATCGGGGGGCTCCCTGCCCTACGCCGATATCGTGAAGG GAAGGTTCACCATTAGCCGGGACAACTCCAAGAACACTCTGTACCTCCA AATGAACAGCCTGAGAGCGGAGGACACCGCAGTGTACTATTGCGCCCGG TACTGGCCAATGGACATCTGGGGCCAGGGGACTCTGGTCACCGTCTCCT CA COMBO_Rd4_0.6nM_C29 light chain variable region (SEQ ID NO: 485) GAGATCGTGCTGACTCAGTCCCCTGGAACCCTGTCCCTGTCACCTGGCG AAAGAGCTACCTTGTCC
  • composition comprises either or both of the polynucleotides shown in SEQ ID NO: 488 and SEQ ID NO: 487 below:
  • composition comprises either or both of the polynucleotides shown in SEQ ID NO: 490 and SEQ ID NO: 489 below:
  • A02_Rd4_0.6nM_C01 heavy chain variable region (SEQ ID NO: 490) GAAGTTCAATTATTGGAATCTGGTGGAGGACTGGTGCAGCCTGGCGGCT CTCTGAGACTGTCTTGTGCCGCCAGCGGCTTCACCTTCAGCAGCTACGC CATGAACTGGGTGCGCCAGGCCCCTGGTAAAGGTTTGGAATGGGTTTCT GCTATTACTGCGTCTGGTGGTTCTACTTACTATGCCGATGTGGTTAAGG GTAGATTCACCATTTCTAGAGACAACTCTAAGAACACCTTGTACTTGCA AATGAACTCCTTGAGAGCTGAAGATACTGCTGTTTATTACTGTGCTAGA TACTGGCCAATGTCGTTGTGGGGTCAAGGTACTCTGGTCACCGTCTCCT CA A02_Rd4_0.6nM_C01 light chain variable region (SEQ ID NO: 489) GAGATCGTGCTGACACAGAGCCCTGGCACCCTGAGCCTGTCTCCTGGTG AAAGAGCTACTTTGTCTTGTAGA
  • composition comprises either or both of the polynucleotides shown in SEQ ID NO: 492 and SEQ ID NO: 491 below:
  • A02_Rd4_0.6nM_C16 heavy chain variable region (SEQ ID NO: 492) GAAGTTCAATTATTGGAATCTGGTGGAGGACTGGTGCAGCCTGGCGGCT CTCTGAGACTGTCTTGTGCCGCCAGCGGCTTCACCTTCAGCAGCTACGC CATGAACTGGGTGCGCCAGGCCCCTGGTAAAGGTTTGGAATGGGTTTCT GCTATTTCTGATTTTGGTGGTTCTACTTACTATGCCGATATCGTTAAGG GTAGATTCACCATTTCTAGAGACAACTCTAAGAACACCTTGTACTTGCA AATGAACTCCTTGAGAGCTGAAGATACTGCTGTTTATTACTGTGCTAGA TACTGGCCAATGGATATTTGGGGTCAAGGTACTCTGGTCACCGTCTCCT CA A02_Rd4_0.6nM_C16 light chain variable region (SEQ ID NO: 491) GAGATCGTGCTGACACAGAGCCCTGGCACCCTGAGCCTGTCTCCTGGTG AAAGAGCTACTTTGTCTTGTAGAGC
  • the invention provides a method of making any of the polynucleotides described herein.
  • Polynucleotides complementary to any such sequences are also encompassed by the present invention.
  • Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules.
  • RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence.
  • Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein.
  • Variants preferably exhibit at least about 70% identity, more preferably, at least about 80% identity, yet more preferably, at least about 90% identity, and most preferably, at least about 95% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M. O., 1978, A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D. G.
  • the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof.
  • Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence).
  • Suitable “moderately stringent conditions” include prewashing in a solution of 5 ⁇ SSC. 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-65° C., 5 ⁇ SSC, overnight; followed by washing twice at 65° C. for 20 minutes with each of 2 ⁇ , 0.5 ⁇ and 0.2 ⁇ SSC containing 0.1% SDS.
  • highly stringent conditions or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5 ⁇ SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 ⁇ Denhardt's solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulf
  • nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
  • a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein.
  • Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome.
  • the polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al., 1989.
  • PCR allows reproduction of DNA sequences.
  • PCR technology is well known in the art and is described in U.S. Pat. Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston, 1994.
  • RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., 1989, supra, for example.
  • Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector.
  • Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, pMB9, CoIE1, pCR1, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
  • Bluescript e.g., pBS SK+
  • shuttle vectors such as pSA3 and pAT28.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462.
  • Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • the vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus).
  • electroporation employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances
  • microprojectile bombardment e.g., where the vector is an infectious agent such as vaccinia virus.
  • infection e.g., where the vector is an infectious agent such as vaccinia virus.
  • the choice of introducing vectors or polynucleotides will often depend on features of the host cell.
  • the invention also provides host cells comprising any of the polynucleotides described herein. Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest.
  • mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462.
  • Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis ) and yeast (such as S. cerevisae, S. pombe ; or K. lactis ).
  • the host cells express the cDNAs at a level of about 5 fold higher, more preferably, 10 fold higher, even more preferably, 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells.
  • Screening the host cells for a specific binding to BCMA or an BCMA domain is effected by an immunoassay or FACS.
  • a cell overexpressing the antibody or protein of interest can be identified.
  • PTA-122094 is a polynucleotide encoding a humanized BCMA antibody heavy chain variable region
  • vector having ATCC Accession No. PTA-122093 is a polynucleotide encoding a humanized BCMA antibody light chain variable region.
  • the present invention also provides a conjugate (or immunoconjugate) of the BCMA antibody as described herein, or of the antigen binding fragment thereof, wherein the antibody or the antigen binding fragment is conjugated to an agent (e.g., a cytotoxic agent) for targeted immunotherapy (e.g., antibody-drug conjugates) either directly or indirectly via a linker.
  • an agent e.g., a cytotoxic agent
  • a cytotoxic agent can be linked or conjugated to the BCMA antibody or the antigen binding fragment thereof as described herein for targeted local delivery of the cytotoxic agent moiety to tumors (e.g., BCMA expressing tumor).
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody (e.g., a carboxyl terminus, an amino terminus, or at another site in the BCMA antibody).
  • the tag comprises an amino acid glutamine (Q) or an amino acid sequence LQG, LLQGG (SEQ ID NO:318), LLQG (SEQ ID NO:454), LSLSQG (SEQ ID NO: 455), GGGLLQGG (SEQ ID NO: 456), GLLQG (SEQ ID NO: 457), LLQ, GSPLAQSHGG (SEQ ID NO: 458), GLLQGGG (SEQ ID NO: 459), GLLQGG (SEQ ID NO: 460), GLLQ (SEQ ID NO: 461), LLQLLQGA (SEQ ID NO: 462), LLQGA (SEQ ID NO: 463), LLQYQGA (SEQ ID NO: 464), LLQGSG (SEQ ID NO: 465), LLQYQG (SEQ ID NO: 466), LLQLLQG (SEQ ID NO: 467), SLLQG (SEQ ID NO: 468), LLQLQ
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody, wherein the tag comprises an amino acid sequence GGLLQGPP (SEQ ID NO: 474) or GGLLQGA (SEQ ID NO: 475) engineered at the light chain carboxyl terminus of the BCMA antibody.
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody, wherein the tag comprises an amino acid sequence LLQG (SEQ ID NO: 454) engineered after residue T135 in the heavy chain of the BCMA antibody.
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody, wherein the tag comprises an amino acid sequence LLQGA (SEQ ID NO: 463) or LLQGPP (SEQ ID NO: 472) engineered at the heavy chain carboxyl terminus of the BCMA antibody and wherein the lysine residue at the heavy chain carboxyl terminus is deleted.
  • the BCMA antibody or the conjugate as described herein comprises an amino acid substitution at position 297 of the BCMA antibody (EU numbering scheme).
  • the amino acid asparagine (N) can be substituted with glutamine (Q) or alanine (A) at position 297 of the BCMA antibody.
  • an isolated antibody comprising an acyl donor glutamine-containing tag and an amino acid modification at position 222, 340, or 370 of the antibody (EU numbering scheme) wherein the modification is an amino acid deletion, insertion, substitution, mutation, or any combination thereof.
  • the BCMA antibody or the conjugate as described herein comprising the acyl donor glutamine-containing tag (e.g., Q, LQG, LLQGG (SEQ ID NO:318), LLQG (SEQ ID NO:454), LSLSQG (SEQ ID NO: 455), GGGLLQGG (SEQ ID NO: 456), GLLQG (SEQ ID NO: 457), LLQ, GSPLAQSHGG (SEQ ID NO: 458), GLLQGGG (SEQ ID NO: 459), GLLQGG (SEQ ID NO: 460), GLLQ (SEQ ID NO: 461), LLQLLQGA (SEQ ID NO: 462), LLQGA (SEQ ID NO: 463), LLQYQGA (SEQ ID NO: 464), LLQGSG (SEQ ID NO: 465), LLQYQG (SEQ ID NO: 466), LLQLLQG (SEQ ID NO:
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence GGLLQGPP (SEQ ID NO: 474) engineered at the C-terminus of the BCMA antibody light chain and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence GGLLQGA (SEQ ID NO: 475) engineered at the C-terminus of the BCMA antibody light chain and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence LLQGA (SEQ ID NO: 463) engineered at the C-terminus of the BCMA antibody heavy chain and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme), wherein the lysine residue at the heavy chain carboxyl terminus is deleted.
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence LLQG (SEQ ID NO: 454) engineered after residue T135 in the heavy chain of the BCMA antibody and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising a glutamine engineered at position 297 or an amino acid substitution at position 297 from asparagine (N) to another amino acid in the BCMA antibody and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence GGLLQGPP (SEQ ID NO: 474) engineered at the C-terminus of the BCMA antibody light chain, an amino acid substitution at position 297 of the BCMA antibody from asparagine (N) to glutamine (Q), and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence LLQG (SEQ ID NO: 454) engineered after residue T135 in the heavy chain of the BCMA antibody, an amino acid substitution at position 297 of the BCMA antibody from asparagine (N) to alanine (A), and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • agents that can be conjugated to the BCMA antibodies or the antigen binding fragments of the present invention include, but are not limited to, cytotoxic agents, immunomodulating agents, imaging agents, therapeutic proteins, biopolymers, or oligonucleotides.
  • cytotoxic agent examples include, but are not limited to, anthracycline, an auristatin, a dolastatin, a combretastatin, a duocarmycin, a pyrrolobenzodiazepine dimer, an indolino-benzodiazepine dimer, an enediyne, a geldanamycin, a maytansine, a puromycin, a taxane, a vinca alkaloid, a camptothecin, a tubulysin, a hemiasterlin, a spliceostatin, a pladienolide, and stereoisomers, isosteres, analogs, or derivatives thereof.
  • the anthracyclines are derived from bacteria Strepomyces and have been used to treat a wide range of cancers, such as leukemias, lymphomas, breast, uterine, ovarian, and lung cancers.
  • exemplary anthracyclines include, but are not limited to, daunorubicin, doxorubicin (i.e., adriamycin), epirubicin, idarubicin, valrubicin, and mitoxantrone.
  • Dolastatins and their peptidic analogs and derivatives, auristatins are highly potent antimitotic agents that have been shown to have anticancer and antifungal activity. See, e.g., U.S. Pat. No. 5,663,149 and Pettit et al., Antimicrob. Agents Chemother. 42:2961-2965, 1998.
  • Exemplary dolastatins and auristatins include, but are not limited to, dolastatin 10, auristatin E, auristatin EB (AEB), auristatin EFP (AEFP), MMAD (Monomethyl Auristatin D or monomethyl dolastatin 10), MMAF (Monomethyl Auristatin F or N-methylvaline-valine-dolaisoleuine-dolaproine-phenylalanine), MMAE (Monomethyl Auristatin E or N-methylvaline-valine-dolaisoleuine-dolaproine-norephedrine), 5-benzoylvaleric acid-AE ester (AEVB), and other novel auristatins (such as the ones described in U.S.
  • the auristatin is 0101 (2-methylalanyl-N-[(3R,4S,5S)-3-methoxy-1- ⁇ (2S)-2-[(1R,2R)-1-methoxy-2-methyl-3-oxo-3- ⁇ [(1 S)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino ⁇ propyl]pyrrolidin-1-yl ⁇ -5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide) having the following structure:
  • the auristatin is 3377 (N,2-dimethylalanyl-N- ⁇ (1 S,2R)-4- ⁇ (2S)-2-[(1R,2R)-3- ⁇ [(1S)-1-carboxyl-2-phenylethyl]amino ⁇ -1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl ⁇ -2-methoxy-1-[(1 S)-1-methylpropyl]-4-oxobutyl ⁇ -N-methyl-L-valinamide) having the following structure:
  • the auristatin is 0131-OMe (N,2-dimethylalanyl-N-[(3R,4S,5S)-3-methoxy-1- ⁇ (2S)-2-[(1R,2R)-1-methoxy-3- ⁇ [(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino ⁇ -2-methyl-3-oxopropyl]pyrrolidin-1-yl ⁇ -5-methyl-1-oxoheptan-4-yl]-N-methylL-valinamide) having the following structure:
  • the auristatin is 0131 (2-methyl-L-prolyl-N-[(3R,4S,5S)-1- ⁇ (2S)-2-[(1R,2R)-3- ⁇ [(1S)-1-carboxy-2-phenylethyl]amino ⁇ -1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl ⁇ -3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide) having the following structure:
  • the auristatin is 0121 (2-methyl-L-proly-N-[(3R,4S,5S)-1- ⁇ (2S)-2-[(1R,2R)-3- ⁇ [(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino ⁇ -1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl ⁇ -3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide) having the following structure:
  • Camptothecin is a cytotoxic quinoline alkaloid which inhibits the enzyme topoisomerase I.
  • Examples of camptothecin and its derivatives include, but are not limited to, topotecan and irinotecan, and their metabolites, such as SN-38.
  • Combretastatins are natural phenols with vascular disruption properties in tumors.
  • Exemplary combretastatins and their derivatives include, but are not limited to, combretastatin A-4 (CA-4) and ombrabulin.
  • Duocarmycin and CC-1065 are DNA alkylating agents with cytotoxic potency. See Boger and Johnson, PNAS 92:3642-3649 (1995).
  • Exemplary duocarmycin and CC-1065 include, but are not limited to, (+)-duocarmycin A and (+)-duocarmycin SA, (+)-CC-1065, and the compounds as disclosed in the international application PCT/IB2015/050280 including, but not limited to, N ⁇ 2 ⁇ acetyl-L-lysyl-L-valyl-N ⁇ 5 ⁇ carbamoyl-N-[4-( ⁇ [(2- ⁇ [( ⁇ (1S)-1-(chloromethyl)-3-[(5- ⁇ [(1S)-1-(chloromethyl)-5-(phosphonooxy)-1,2-dihydro-3H-benzo[e]indol-3-yl]carbonyl ⁇ thiophen-2-yl)carbonyl]-2,3-dihydro-1
  • Enediynes are a class of anti-tumor bacterial products characterized by either nine- and ten-membered rings or the presence of a cyclic system of conjugated triple-double-triple bonds.
  • Exemplary enediynes include, but are not limited to, calicheamicin, esperamicin, uncialamicin, dynemicin, and their derivatives.
  • Geldanamycins are benzoquinone ansamycin antibiotic that bind to Hsp90 (Heat Shock Protein 90) and have been used antitumor drugs.
  • exemplary geldanamycins include, but are not limited to, 17-AAG (17-N-Allylamino-17-Demethoxygeldanamycin) and 17-DMAG (17-Dimethylaminoethylamino-17-demethoxygeldanamycin).
  • Hemiasterlin and its analogues bind to the tubulin, disrupt normal microtubule dynamics, and, at stoichiometric amounts, depolymerize microtubules.
  • Maytansines or their derivatives maytansinoids inhibit cell proliferation by inhibiting the microtubules formation during mitosis through inhibition of polymerization of tubulin. See Remillard et al., Science 189:1002-1005, 1975.
  • Exemplary maytansines and maytansinoids include, but are not limited to, mertansine (DM1) and its derivatives as well as ansamitocin.
  • PBDs Pyrrolobenzodiazepine dimers
  • IGNs indolino-benzodiazepine dimers
  • PBD and IGN molecules are based on the natural product athramycin, and interact with DNA in a sequence-selective manner, with a preference for purine-guanine-purine sequences.
  • Exemplary PBDs and their analogs include, but are not limited to, SJG-136.
  • Spliceostatins and pladienolides are anti-tumor compounds which inhibit splicing and interacts with spliceosome, SF3b.
  • spliceostatins include, but are not limited to, spliceostatin A, FR901464, and (2S,3Z)-5- ⁇ [(2R,3R,5S,6S)-6- ⁇ (2E,4E)-5-[(3R,4R,5R,7S)-4-hydroxy-1,6-dioxaspiro[2.5]oct-5-yl]-3-methylpenta-2,4-dien-1-yl ⁇ -2,5-dimethyltetrahydro-2H-pyran-3-yl]amino ⁇ -5-oxopent-3-en-2-yl acetate having the structure of
  • pladienolides examples include, but are not limited to, Pladienolide B, Pladienolide D, or E7107.
  • Taxanes are diterpenes that act as anti-tubulin agents or mitotic inhibitors.
  • Exemplary taxanes include, but are not limited to, paclitaxel (e.g., TAXOL®) and docetaxel (TAXOTERE®).
  • Tubulysins are natural products isolated from a strain of myxobacteria that has been shown to depolymerize microtubules and induce mitotic arrest.
  • Exemplary tubulysins include, but are not limited to, tubulysin A, tubulysin B, and tubulysin D.
  • Vinca alkyloids are also anti-tubulin agents.
  • Exemplary vinca alkyloids include, but are not limited to, vincristine, vinblastine, vindesine, and vinorelbine.
  • the cytotoxic agent is selected from the group consisting of MMAD (Monomethyl Auristatin D), 0101 (2-methylalanyl-N-[(3R,4S,5S)-3-methoxy-1- ⁇ (2S)-2-[(1R,2R)-1-methoxy-2-methyl-3-oxo-3- ⁇ [(1 S)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino ⁇ propyl]pyrrolidin-1-yl ⁇ -5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), 3377 (N,2-dimethylalanyl-N- ⁇ (1 S,2R)-4- ⁇ (2S)-2-[(1R,2R)-3- ⁇ [(1 S)-1-carboxyl-2-phenylethyl]amino ⁇ -1-methoxy-2-methyl-3-oxopropyl]pyrroidin-1
  • MMAD Mono
  • the agent is an immunomodulating agent.
  • an immunomodulating agent include, but are not limited to, gancyclovier, etanercept, tacrolimus, sirolimus, voclosporin, cyclosporine, rapamycin, cyclophosphamide, azathioprine, mycophenolgate mofetil, methotrextrate, glucocorticoid and its analogs, cytokines, stem cell growth factors, lymphotoxins, tumor necrosis factor (TNF), hematopoietic factors, interleukins (e.g., interleukin-1 (IL-1), IL-2, IL-3, IL-6, IL-10, IL-12, IL-18, and IL-21), colony stimulating factors (e.g., granulocyte-colony stimulating factor (G-CSF) and granulocyte macrophage-colony stimulating factor (GM-CSF)), interferons (e.g., interferons- ⁇
  • the agent moiety is an imaging agent (e.g., a fluorophore or a chelator), such as fluorescein, rhodamine, lanthanide phosphors, and their derivatives thereof, or a radioisotope bound to a chelator.
  • an imaging agent e.g., a fluorophore or a chelator
  • fluorescein, rhodamine, lanthanide phosphors, and their derivatives thereof, or a radioisotope bound to a chelator e.g., a fluorophore or a chelator
  • fluorophores examples include, but are not limited to, fluorescein isothiocyanate (FITC) (e.g., 5-FITC), fluorescein amidite (FAM) (e.g., 5-FAM), eosin, carboxyfluorescein, erythrosine, Alexa Fluor® (e.g., Alexa 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 647, 660, 680, 700, or 750), carboxytetramethylrhodamine (TAMRA) (e.g., 5-TAMRA), tetramethylrhodamine (TMR), and sulforhodamine (SR) (e.g., SR101).
  • FITC fluorescein isothiocyanate
  • FAM fluorescein amidite
  • eosin carboxyfluorescein, erythrosine
  • chelators include, but are not limited to, 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic acid (DOTA), 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (deferoxamine), diethylenetriaminepentaacetic acid (DTPA), and 1,2-bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid) (BAPTA).
  • DOTA 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic acid
  • NOTA 1,4,7-triazacyclononane-1,4,7-triacetic acid
  • DTPA diethylenetriaminepentaacetic acid
  • BAPTA 1,2-bis(o-aminophen
  • fluorophores examples include, but are not limited to, fluorescein isothiocyanate (FITC) (e.g., 5-FITC), fluorescein amidite (FAM) (e.g., 5-FAM), eosin, carboxyfluorescein, erythrosine, Alexa Fluor® (e.g., Alexa 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 647, 660, 680, 700, or 750), carboxytetramethylrhodamine (TAMRA) (e.g., 5-TAMRA), tetramethylrhodamine (TMR), and sulforhodamine (SR) (e.g., SR101).
  • FITC fluorescein isothiocyanate
  • FAM fluorescein amidite
  • eosin carboxyfluorescein, erythrosine
  • therapeutic or diagnostic radioisotopes or other labels can be incorporated in the agent for conjugation to the BCMA antibodies or the antigen binding fragments as described herein.
  • a radioisotope or other labels include, but are not limited to, 3 H, 11 C, 13 N, 14 C, 15 N, 15 O, 35 S, 18 F 32 P, 33 P, 47 Sc, 51 Cr, 57 Co, 58 Co, 59 Fe, 62 Cu, 64 Cu, 67 Cu, 67 Ga, 68 Ga, 75 Se, 76 Br, 77 Br, 86 Y 89 Zr, 90 Y, 94 Tc, 95 Ru, 97 Ru, 99 Tc, 103 Ru, 105 Rh, 105 Ru, 107 Hg, 109 Pd, 111 Ag, 111 In, 113 In, 121 Te, 122 Te, 123 I, 124 I, 125 I, 125 Te, 126 I, 131 I, 131 In, 133
  • the agent is a therapeutic protein including, but is not limited to, a toxin, a hormone, an enzyme, and a growth factor.
  • toxin protein examples include, but are not limited to, dipththeria (e.g., diphtheria A chain), Pseudomonas exotoxin and endotoxin, ricin (e.g., ricin A chain), abrin (e.g., abrin A chain), modeccin (e.g., modeccin A chain), alpha-sarcin, Aleurites fordii proteins, dianthin proteins, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtherin toxin, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), Momordica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, mitogellin, restrictocin, phenomycin, enomycin, tricothecenes, inhibitor cyst
  • the agent is a biocompatible polymer.
  • the BCMA antibodies or the antigen binding fragments as described herein can be conjugated to the biocompatible polymer to increase serum half-life and bioactivity, and/or to extend in vivo half-lives.
  • biocompatible polymers include water-soluble polymer, such as polyethylene glycol (PEG) or its derivatives thereof and zwitterion-containing biocompatible polymers (e.g., a phosphorylcholine containing polymer).
  • the agent is an oligonucleotide, such as anti-sense oligonucleotides.
  • the invention provides a conjugate of the antibody or the antigen binding fragment as described herein, wherein the conjugate comprises the formula: antibody-(acyl donor glutamine-containing tag)-(linker)-(cytotoxic agent), wherein the acyl donor glutamine-containing tag is engineered at a specific site of the antibody or the antigen binding fragment (e.g., at a carboxyl terminus of the heavy or light chain, after residue T135 in the antibody heavy chain, or at an another site), wherein the tag is conjugated to a linker (e.g., a linker containing one or more reactive amines (e.g., primary amine NH2)), and wherein the linker is conjugated to a cytotoxic agent (e.g., MMAD or other auristatins such as 0101, 0131, or 3377).
  • a linker e.g., a linker containing one or more reactive amines (e.g., primary amine NH2)
  • a cytotoxic agent
  • Examples of a linker containing one or more reactive amines include, but are not limited to, Ac-Lys-Gly (acetyl-lysine-glycine), aminocaproic acid, Ac-Lys- ⁇ -Ala (acetyl-lysine- ⁇ -alanine), amino-PEG2 (polyethylene glycol)-C2, amino-PEG3-C2, amino-PEG6-C2 (or amino PEG6-propionyl), Ac-Lys-Val-Cit-PABC (acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl), amino-PEG6-C2-Val-Cit-PABC, aminocaproyl-Val-Cit-PABC, [(3R,5R)-1- ⁇ 3-[2-(2-aminoethoxy)ethoxy]propanoyl ⁇ piperidine-3,5-diyl]bis-Val-Cit-PABC, [(3S,
  • the conjugate is 1) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101; 2) antibody-AcLys-VC-PABC-0101 and comprises N297Q; 3) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101 and comprises N297Q; 4) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-0131 and comprises N297A; 5) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-3377 and comprises N297A; 6) antibody-GGLLQGA (SEQ ID NO: 475)-AcLys-VC-PABC-0101.
  • the acyl donor glutamine-containing tag comprising, e.g., GGLLQGPP (SEQ ID NO: 474) or GGLLQGA (SEQ ID NO: 475), is engineered at the C-terminus of the light chain of the antibody.
  • the acyl donor glutamine-containing tag e.g., LLQGA (SEQ ID NO: 463) or LLQGPP (SEQ ID NO: 472)
  • the acyl donor glutamine-containing tag is engineered at the C-terminus of the heavy chain of the antibody, wherein the lysine residue at the C-terminus is deleted.
  • the acyl donor glutamine-containing tag comprising, e.g., LLQG (SEQ ID NO: 454) is engineered after residue T135 in the antibody heavy chain or replaces amino acid residues E294-N297 in the antibody heavy chain.
  • Examples of the antibody include, but are not limited to, P6E01/P6E01, P6E01/H3.AQ, L1.LGF/L3.KW/P6E01; L1.LGF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.LGF/L3.KW/H3.AL, L1.LGF/L3.KW/H3.AP, L1.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.NY/H3.AL, L1.GDF/L3.NY/
  • the conjugate further comprises an amino acid substitution from lysine to arginine at position 222.
  • the conjugate is 1) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101 and comprises K222R; 2) antibody-AcLys-VC-PABC-0101 and comprises N297Q and K222R; 3) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101 and comprises N297Q and K222R; 4) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-0131 and comprises N297A and K222R; 5) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-3377 and comprises N297A and K222R; and 6) antibody-GGLLQGA (SEQ ID NO: 475)-AcLys-VC-PABC-0101 and comprises K222R.
  • the acyl donor glutamine-containing tag comprising, e.g., GGLLQGPP (SEQ ID NO: 474) or GGLLQGA (SEQ ID NO: 475) is engineered at the C-terminus of the light chain of the antibody.
  • the acyl donor glutamine-containing tag e.g., LLQGA (SEQ ID NO: 473) or LLQGPP (SEQ ID NO: 472)
  • the acyl donor glutamine-containing tag is engineered at the C-terminus of the heavy chain of the antibody, wherein the lysine residue at the C-terminus is deleted.
  • the acyl donor glutamine-containing tag comprising, e.g., LLQG (SEQ ID NO: 454) is engineered after residue T135 in the antibody heavy chain or replaces amino acid residues E294-N297 in the antibody heavy chain.
  • Examples of the antibody include, but are not limited to, P6E01/P6E01, P6E01/H3.AQ, L1.LGF/L3.KW/P6E01, L1.LGF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.LGF/L3.KW/H3.AL, L1.LGF/L3.KW/H3.AP, L1.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.NY
  • the present invention further provides an antibody that binds to CD3 (e.g., human CD3 (SEQ ID NO: 502; or accession number: NM_000733.3).
  • CD3 e.g., human CD3 (SEQ ID NO: 502; or accession number: NM_000733.3).
  • VL light chain variable
  • VL light chain variable
  • an antibody having any one of partial light chain sequence as listed in Table 3 and/or any one of partial heavy chain sequence as listed in Table 3.
  • the invention also provides CDR portions of antibodies to CD3 (including Chothia, Kabat CDRs, and CDR contact regions). Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed “combined CRs” or “extended CDRs”). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof. Table 4 provides examples of CDR sequences provided herein.
  • the invention also provides isolated polynucleotides encoding the antibodies of the invention, and vectors and host cells comprising the polynucleotide.
  • a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody h2B4, h2B4-VH-wt VL_TK, h2B4-VH-hnps VL_TK, h2B4-VH-yaes VL_TK, h2B4-VH-yads VL_TK, h2B4-VH-yaps VL_TK, h2B4-VH-hnps VL_TK-S55Y, h2B4-VH-hnps VL_TK-S105Q, h2B4-vH-hnps VL_TK-S55Y/S105Q, 2B34, h21B4-11, 1010, 1A4, 7A3, 25A8, 16G7, h25A8-B5, h25A8-B8, h25A8-B12, h25A8-B
  • fusion proteins comprising one or more fragments or regions from the antibodies of this invention.
  • a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of the variable light chain region shown in SEQ ID NOs: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 445, 352, 355, 443, 377, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399, and/or at least 10 amino acids of the variable heavy chain region shown in SEQ ID NOs: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 354, 356, 444, 442, 378, 380, 382, 384, 386, 388, 390, 392, 394, 396, 398, or 400.
  • a fusion polypeptide comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region.
  • the fusion polypeptide comprises a light chain variable region and/or a heavy chain variable region, as shown in any of the sequence pairs selected from among SEQ ID NOs: 319 and 320, 321 and 322, 323 and 324, 325 and 326, 327 and 328, 329 and 330, 344 and 345, 346 and 347, 348 and 349, 350 and 351, 445 and 444, 352 and 354, 355 and 356, 443 and 442, 377 and 378, 379 and 380, 381 and 382, 383 and 384, 385 and 386, 387 and 388, 389 and 390, 391 and 392, 393 and 394, 395 and 396, 397 and 398, or 399 and 400.
  • the fusion polypeptide comprises one or more CDR(s).
  • the fusion polypeptide comprises CDR H3 (VH CDR3) and/or CDR L3 (VL CDR3).
  • a fusion protein contains one or more antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region.
  • heterologous sequences include, but are not limited to a “tag” such as a FLAG tag or a 6His tag. Tags are well known in the art.
  • a fusion polypeptide can be created by methods known in the art, for example, synthetically or recombinantly.
  • the fusion proteins of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, although they may also be prepared by other means known in the art, including, for example, chemical synthesis.
  • CD3 antibody in the present invention Representative materials of the CD3 antibody in the present invention were deposited in the American Type Culture Collection (ATCC) on Sep. 11, 2015.
  • Vector having ATCC Accession No. PTA-122513 is a polynucleotide encoding a humanized CD3 antibody heavy chain variable region
  • vector having ATCC Accession No. PTA-122512 is a polynucleotide encoding a humanized CD3 antibody light chain variable region.
  • the deposits were made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and Regulations thereunder (Budapest Treaty). This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit.
  • Bispecific antibodies monoclonal antibodies that have binding specificities for at least two different antigens
  • Methods for making bispecific antibodies are known in the art (see, e.g., Suresh et al., Methods in Enzymology 121:210, 1986).
  • the recombinant production of bispecific antibodies was based on the coexpression of two immunoglobulin heavy chain-light chain pairs, with the two heavy chains having different specificities (Millstein and Cuello, Nature 305, 537-539, 1983).
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant region sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant region, comprising at least part of the hinge, CH2 and CH3 regions. It is preferred to have the first heavy chain constant region (CH1), containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are cotransfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm.
  • This asymmetric structure with an immunoglobulin light chain in only one half of the bispecific molecule, facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations. This approach is described in PCT Publication No. WO 94/04690.
  • the bispecific antibodies are composed of amino acid modification in the first hinge region in one arm, and the substituted/replaced amino acid in the first hinge region has an opposite charge to the corresponding amino acid in the second hinge region in another arm.
  • This approach is described in International Patent Application No. PCT/US2011/036419 (WO2011/143545).
  • a desired heteromultimeric or heterodimeric protein is enhanced by altering or engineering an interface between a first and a second immunoglobulin-like Fc region (e.g., a hinge region and/or a CH3 region).
  • the bispecific antibodies may be composed of a CH3 region, wherein the CH3 region comprises a first CH3 polypeptide and a second CH3 polypeptide which interact together to form a CH3 interface, wherein one or more amino acids within the CH3 interface destabilize homodimer formation and are not electrostatically unfavorable to homodimer formation.
  • the bispecific antibodies can be generated using a glutamine-containing peptide tag engineered to the antibody directed to an epitope (e.g., BCMA) in one arm and another peptide tag (e.g., a Lys-containing peptide tag or a reactive endogenous Lys) engineered to a second antibody directed to a second epitope in another arm in the presence of transglutaminase.
  • an epitope e.g., BCMA
  • another peptide tag e.g., a Lys-containing peptide tag or a reactive endogenous Lys
  • the heterodimeric protein (e.g., bispecific antibody) as described herein comprises a full-length human antibody, wherein a first antibody variable domain of the heterodimeric protein is capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen located on the human immune effector cell, and wherein a second antibody variable domain of the heterodimeric protein is capable of specifically binding to a target antigen.
  • the human antibody has an IgG1, IgG2, IgG3, or IgG4 isotype.
  • the heterodimeric protein comprises an immunologically inert Fc region.
  • the human immune effector cell can be any of a variety of immune effector cells known in the art.
  • the immune effector cell can be a member of the human lymphoid cell lineage, including, but not limited to, a T cell (e.g., a cytotoxic T cell), a B cell, and a natural killer (NK) cell.
  • the immune effector cell can also be, for example without limitation, a member of the human myeloid lineage, including, but not limited to, a monocyte, a neutrophilic granulocyte, and a dendritic cell.
  • Such immune effector cells may have either a cytotoxic or an apoptotic effect on a target cell or other desired effect upon activation by binding of an effector antigen.
  • the effector antigen is an antigen (e.g., a protein or a polypeptide) that is expressed on the human immune effector cell.
  • antigens e.g., a protein or a polypeptide
  • effector antigens that can be bound by the heterodimeric protein include, but are not limited to, human CD3 (or CD3 (Cluster of Differentiation) complex), CD16, NKG2D, NKp46, CD2, CD28, CD25, CD64, and CD89.
  • the target cell can be a cell that is native or foreign to humans.
  • the cell may have been transformed to be a malignant cell or pathologically modified (e.g., a native target cell infected with a virus, a plasmodium , or a bacterium).
  • a native target cell the cell is an invading pathogen, such as a bacterium, a plasmodium , or a virus.
  • the target antigen is expressed on a target cell in a diseased condition (e.g., an inflammatory disease, a proliferative disease (e.g., cancer), an immunological disorder, a neurological disease, a neurodegenerative disease, an autoimmune disease, an infectious disease (e.g., a viral infection or a parasitic infection), an allergic reaction, a graft-versus-host disease or a host-versus-graft disease).
  • a diseased condition e.g., an inflammatory disease, a proliferative disease (e.g., cancer), an immunological disorder, a neurological disease, a neurodegenerative disease, an autoimmune disease, an infectious disease (e.g., a viral infection or a parasitic infection), an allergic reaction, a graft-versus-host disease or a host-versus-graft disease).
  • a target antigen is not effector antigen.
  • target antigens include, but are not limited to, BCMA, EpCAM (Epithelial Cell Adhesion Molecule), CCR5 (Chemokine Receptor type 5), CD19, HER (Human Epidermal Growth Factor Receptor)-2/neu, HER-3, HER-4, EGFR (Epidermal Growth Factor Receptor), PSMA, CEA, MUC-1 (Mucin), MUC2, MUC3, MUC4, MUCSAC, MUC5B, MUC7, ClhCG, Lewis-Y, CD20, CD33, CD30, ganglioside GD3, 9-O-Acetyl-GD3, GM2, Globo H, fucosyl GM1, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Shh (Sonic Hedgehog), Wue-1, Plasma Cell Antigen, (membrane-bound) IgE, MCSP (Melanoma Chondroitin Sulfate
  • the heterodimeric protein (e.g., bispecific antibody) as described herein comprises a full-length human antibody, wherein a first antibody variable domain of the heterodimeric protein is capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen (e.g., CD3 antigen) located on the human immune effector cell, wherein a second antibody variable domain of the heterodimeric protein is capable of specifically binding to a target antigen (e.g., CD20 antigen or EpCAM), wherein the first and second antibody variable domain of the heterodimeric protein comprise amino acid modifications at positions 223, 225, and 228 (e.g., (C223E or C223R), (E225R), and (P228E or P228R)) in the hinge region and at position 409 or 368 (e.g., K409R or L368E (EU numbering scheme)) in the CH3 region of human IgG2 (SEQ ID NO: 493).
  • an effector antigen
  • the first and second antibody variable domains of the heterodimeric protein comprise amino acid modifications at positions 221 and 228 (e.g., (D221R or D221E) and (P228R or P228E)) in the hinge region and at position 409 or 368 (e.g., K409R or L368E (EU numbering scheme)) in the CH3 region of human IgG1 (SEQ ID NO: 494).
  • positions 221 and 228 e.g., (D221R or D221E) and (P228R or P228E)
  • position 409 or 368 e.g., K409R or L368E (EU numbering scheme)
  • the first and second antibody variable domains of the heterodimeric protein comprise amino acid modifications at positions 228 (e.g., (P228E or P228R)) in the hinge region and at position 409 or 368 (e.g., R409 or L368E (EU numbering scheme)) in the CH3 region of human IgG4 (SEQ ID NO: 495).
  • positions 228 e.g., (P228E or P228R)
  • position 409 or 368 e.g., R409 or L368E (EU numbering scheme)
  • the first antibody variable domain of the heterodimeric protein comprises a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443, 445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399, and the second antibody variable domain of the heterodimeric protein comprises VH region comprising VH C
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387; and the second antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 112; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 38.
  • VH heavy chain variable
  • VL light chain variable
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 417; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 156; (ii) a VH C
  • the VH CDR1 of the first antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 331.
  • the VH CDR1 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 333.
  • the VH CDR2 of the first antibody variable domain may further comprise the sequence of F or FI immediately preceding the sequence shown in SEQ ID NO: 417 and/or the sequence of S, SD, SDH, SDHN, SDHNP, SDHNPS, SDHNPSV, SDHNPSVK, or SDHNPSVKG immediately following the sequence shown in SEQ ID NO: 417.
  • the VH CDR2 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 336.
  • the VH CDR1 of the second antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 156.
  • the VH CDR1 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 157.
  • the VH CDR2 of the second antibody variable domain may further comprise the sequence of A or AI immediately preceding the sequence shown in SEQ ID NO: 159 and/or the sequence of L, LP, LPY, LPYA, LPYAD, LPYADS, LPYADSV, LPYADSVK, or LPYADSVKG immediately following the sequence shown in SEQ ID NO: 159.
  • the VH CDR2 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 158.
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 332 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 151; (ii) a VH C
  • the VH CDR1 of the first antibody variable domain may further comprise the sequence of Y, YM or YMT immediately following the sequence shown in SEQ ID NO: 332.
  • the VH CDR1 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 333.
  • the VH CDR1 of the second antibody variable domain may further comprise the sequence P, PM, or PMS immediately following the sequence shown in SEQ ID NO: 151.
  • the VH CDR1 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 157.
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 401 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 404; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 406; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 439; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 156; (ii) a VH C
  • the VH CDR1 of the first antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 401.
  • the VH CDR1 of the first antibody variable domain comprises the sequence shown in SEQ ID NO: 408.
  • the VH CDR2 of the first antibody variable domain may further comprise the sequence of R or RI immediately preceding the sequence shown in SEQ ID NO: 404 and/or the sequence of T, TY, TYY, TYYA, TYYAE, TYYAES, TYYAESV, TYYAESVK, or TYYAESVKG immediately following the sequence shown in SEQ ID NO: 404.
  • the bispecific antibody of claim 49 wherein the VH CDR2 of the first antibody variable domain comprises the sequence shown in SEQ ID NO: 405.
  • the VH CDR1 of the second antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 156.
  • the VH CDR1 of the second antibody variable domain comprises the sequence shown in SEQ ID NO: 157.
  • the VH CDR2 of the second antibody variable domain may further comprise the sequence of A or AI immediately preceding the sequence shown in SEQ ID NO: 159 and/or the sequence of L, LP, LPY, LPYA, LPYAD, LPYADS, LPYADSV, LPYADSVK, or LPYADSVKG immediately following the sequence shown in SEQ ID NO: 159.
  • the VH CDR2 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 158.
  • the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 407 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 405; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 406; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 439; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 151; (ii) a VH C
  • the VH CDR1 of the first antibody variable domain may further comprise the sequence of A, AM or AMN immediately following the sequence shown in SEQ ID NO: 407.
  • the VH CDR1 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 408.
  • the VH CDR1 of the second antibody variable domain may further comprise the sequence P, PM, or PMS immediately following the sequence shown in SEQ ID NO: 151.
  • the VH CDR1 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 157.
  • the antibodies useful in the present invention can encompass monoclonal antibodies, polyclonal antibodies, antibody fragments (e.g., Fab, Fab′, F(ab′)2, Fv, Fc, etc.), chimeric antibodies, bispecific antibodies, heteroconjugate antibodies, single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion (e.g., a domain antibody), humanized antibodies, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies.
  • the antibodies may be murine, rat, human, or any other origin (including chimeric or humanized antibodies).
  • the BCMA or CD3 antibody as described herein is a monoclonal antibody.
  • the BCMA or CD3 antibody is a humanized monoclonal antibody or a chimeric monoclonal antibody.
  • the antibody comprises a modified constant region, such as, for example without limitation, a constant region that has increased potential for provoking an immune response.
  • the constant region may be modified to have increased affinity to an Fc gamma receptor such as, e.g., Fc ⁇ RI, Fc ⁇ RIIA, or Fc ⁇ III.
  • the antibody comprises a modified constant region, such as a constant region that is immunologically inert, that is, having a reduced potential for provoking an immune response.
  • the constant region is modified as described in Eur. J. Immunol., 29:2613-2624, 1999; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 98099518.
  • the Fc can be human IgG1, human IgG2, human IgG3, or human IgG4.
  • the Fc can be human IgG2 containing the mutation A330P331 to S330S331 (IgG2Aa), in which the amino acid residues are numbered with reference to the wild type IgG2 sequence.
  • the antibody comprises a constant region of IgG4 comprising the following mutations (Armour et al., Molecular Immunology 40 585-593, 2003): E233F234L235 to P233V234A235 (IgG4Ac), in which the numbering is with reference to wild type IgG4.
  • the Fc is human IgG4 E233F234L235 to P233V234A235 with deletion G236 (IgG4Ab).
  • the Fc is any human IgG4 Fc (IgG4, IgG4Ab or IgG4Ac) containing hinge stabilizing mutation S228 to P228 (Aalberse et al., Immunology 105, 9-19, 2002).
  • the Fc can be aglycosylated Fc.
  • the constant region is aglycosylated by mutating the oligosaccharide attachment residue (such as Asn297) and/or flanking residues that are part of the glycosylation recognition sequence in the constant region.
  • the constant region is aglycosylated for N-linked glycosylation enzymatically.
  • the constant region may be aglycosylated for N-linked glycosylation enzymatically or by expression in a glycosylation deficient host cell.
  • the constant region has a modified constant region that removes or reduces Fc gamma receptor binding.
  • the Fc can be human IgG2 containing the mutation D265, in which the amino acid residues are numbered with reference to the wild type IgG2 sequence (SEQ ID NO: 493).
  • the constant region has a modified constant region having the sequence shown in SEQ ID NO: 496:
  • the constant region has a modified constant region having the sequence shown in SEQ ID NO: 497:
  • One way of determining binding affinity of antibodies to BCMA or CD3 is by measuring binding affinity of monofunctional Fab fragments of the antibody.
  • an antibody for example, IgG
  • an antibody can be cleaved with papain or expressed recombinantly.
  • the affinity of a BCMA Fab fragment of an antibody can be determined by surface plasmon resonance (BiacoreTM3000TM surface plasmon resonance (SPR) system, BiacoreTM, INC, Piscataway NJ) equipped with pre-immobilized streptavidin sensor chips (SA) or anti-mouse Fc or anti-human Fc using HBS-EP running buffer (0.01M HEPES, pH 7.4, 0.15 NaCl, 3 mM EDTA, 0.005% v/v Surfactant P20).
  • surface plasmon resonance BiacoreTM3000TM surface plasmon resonance (SPR) system, BiacoreTM, INC, Piscataway NJ
  • SA pre-immobilized streptavidin sensor chips
  • HBS-EP running buffer 0.01M HEPES, pH 7.4, 0.15 NaCl, 3 mM EDTA, 0.005% v/v Surfactant P20.
  • Biotinylated or Fc fusion human BCMA can be diluted into HBS-EP buffer to a concentration of less than 0.5 pg/mL and injected across the individual chip channels using variable contact times, to achieve two ranges of antigen density, either 50-200 response units (RU) for detailed kinetic studies or 800-1,000 RU for screening assays.
  • Regeneration studies have shown that 25 mM NaOH in 25% v/v ethanol effectively removes the bound Fab while keeping the activity of BCMA on the chip for over 200 injections.
  • serial dilutions spanning concentrations of 0.1-10 ⁇ estimated K D
  • purified Fab samples are injected for 1 min at 100 ⁇ L/minute and dissociation times of up to 2 hours are allowed.
  • the concentrations of the Fab proteins are determined by ELISA and/or SDS-PAGE electrophoresis using a Fab of known concentration (as determined by amino acid analysis) as a standard.
  • Kinetic association rates (k on ) and dissociation rates (k off ) are obtained simultaneously by fitting the data globally to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program.
  • Equilibrium dissociation constant (K D ) values are calculated as k off /k on .
  • This protocol is suitable for use in determining binding affinity of an antibody to any BCMA, including human BCMA, BCMA of another mammal (such as mouse BCMA, rat BCMA, or primate BCMA), as well as different forms of BCMA (e.g., glycosylated BCMA). Binding affinity of an antibody is generally measured at 25° C., but can also be measured at 37° C.
  • the antibodies as described herein may be made by any method known in the art.
  • the route and schedule of immunization of the host animal are generally in keeping with established and conventional techniques for antibody stimulation and production, as further described herein.
  • General techniques for production of human and mouse antibodies are known in the art and/or are described herein.
  • any mammalian subject including humans or antibody producing cells therefrom can be manipulated to serve as the basis for production of mammalian, including human and hybridoma cell lines.
  • the host animal is inoculated intraperitoneally, intramuscularly, orally, subcutaneously, intraplantar, and/or intradermally with an amount of immunogen, including as described herein.
  • Hybridomas can be prepared from the lymphocytes and immortalized myeloma cells using the general somatic cell hybridization technique of Kohler, B. and Milstein, C., Nature 256:495-497, 1975 or as modified by Buck, D. W., et al., In Vitro, 18:377-381, 1982.
  • Available myeloma lines including but not limited to X63-Ag8.653 and those from the Salk Institute, Cell Distribution Center, San Diego, Calif., USA, may be used in the hybridization.
  • the technique involves fusing myeloma cells and lymphoid cells using a fusogen such as polyethylene glycol, or by electrical means well known to those skilled in the art.
  • the cells are separated from the fusion medium and grown in a selective growth medium, such as hypoxanthine-aminopterin-thymidine (HAT) medium, to eliminate unhybridized parent cells.
  • a selective growth medium such as hypoxanthine-aminopterin-thymidine (HAT) medium
  • HAT hypoxanthine-aminopterin-thymidine
  • Any of the media described herein, supplemented with or without serum, can be used for culturing hybridomas that secrete monoclonal antibodies.
  • EBV immortalized B cells may be used to produce the monoclonal antibodies of the subject invention.
  • hybridomas are expanded and subcloned, if desired, and supernatants are assayed for anti-immunogen activity by conventional immunoassay procedures (e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay).
  • immunoassay procedures e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay.
  • Hybridomas that may be used as source of antibodies encompass all derivatives, progeny cells of the parent hybridomas that produce monoclonal antibodies specific for BCMA, CD3, or portions thereof.
  • Hybridomas that produce such antibodies may be grown in vitro or in vivo using known procedures.
  • the monoclonal antibodies may be isolated from the culture media or body fluids, by conventional immunoglobulin purification procedures such as ammonium sulfate precipitation, gel electrophoresis, dialysis, chromatography, and ultrafiltration, if desired.
  • Undesired activity, if present, can be removed, for example, by running the preparation over adsorbents made of the immunogen attached to a solid phase and eluting or releasing the desired antibodies off the immunogen.
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin,
  • the antibody (monoclonal or polyclonal) of interest may be sequenced and the polynucleotide sequence may then be cloned into a vector for expression or propagation.
  • the sequence encoding the antibody of interest may be maintained in vector in a host cell and the host cell can then be expanded and frozen for future use.
  • Production of recombinant monoclonal antibodies in cell culture can be carried out through cloning of antibody genes from B cells by means known in the art. See, e.g. Tiller et al., J. Immunol. Methods 329, 112, 2008; U.S. Pat. No. 7,314,622.
  • the polynucleotide sequence may be used for genetic manipulation to “humanize” the antibody or to improve the affinity, or other characteristics of the antibody.
  • the constant region may be engineered to more nearly resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. It may be desirable to genetically manipulate the antibody sequence to obtain greater affinity to BCMA or CD3 and greater efficacy in inhibiting BCMA.
  • a number of “humanized” antibody molecules comprising an antigen binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent or modified rodent V regions and their associated CDRs fused to human constant regions. See, for example, Winter et al. Nature 349:293-299, 1991, Lobuglio et al. Proc. Nat. Acad. Sci. USA 86:4220-4224, 1989, Shaw et al. J Immunol. 138:4534-4538, 1987, and Brown et al. Cancer Res. 47:3577-3583, 1987. Other references describe rodent CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody constant region.
  • FR human supporting framework region
  • humanized antibodies discussed above are also applicable to customizing antibodies for use, for example, in dogs, cats, primate, equines and bovines. Further, one or more aspects of humanizing an antibody described herein may be combined, e.g., CDR grafting, framework mutation and CDR mutation.
  • Fully human antibodies may be obtained by using commercially available mice that have been engineered to express specific human immunoglobulin proteins.
  • Transgenic animals that are designed to produce a more desirable (e.g., fully human antibodies) or more robust immune response may also be used for generation of humanized or human antibodies. Examples of such technology are XenomouseTM from Abgenix, Inc. (Fremont, CA) and HuMAb-Mouse® and TC MouseTM from Medarex, Inc. (Princeton, NJ).
  • antibodies may be made recombinantly and expressed using any method known in the art.
  • antibodies may be made recombinantly by phage display technology. See, for example, U.S. Pat. Nos. 5,565,332; 5,580,717; 5,733,743; and 6,265,150; and Winter et al., Annu. Rev. Immunol. 12:433-455, 1994.
  • the phage display technology (McCafferty et al., Nature 348:552-553, 1990) can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
  • V immunoglobulin variable
  • antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle.
  • a filamentous bacteriophage such as M13 or fd
  • the filamentous particle contains a single-stranded DNA copy of the phage genome
  • selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage mimics some of the properties of the B cell.
  • Phage display can be performed in a variety of formats; for review see, e.g., Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3:564-571, 1993.
  • V-gene segments can be used for phage display. Clackson et al., Nature 352:624-628, 1991, isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice. A repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Mark et al., J. Mol. Biol. 222:581-597, 1991, or Griffith et al., EMBO J. 12:725-734, 1993.
  • Antibodies may be made recombinantly by first isolating the antibodies and antibody producing cells from host animals, obtaining the gene sequence, and using the gene sequence to express the antibody recombinantly in host cells (e.g., CHO cells). Another method which may be employed is to express the antibody sequence in plants (e.g., tobacco) or transgenic milk. Methods for expressing antibodies recombinantly in plants or milk have been disclosed. See, for example, Peeters, et al. Vaccine 19:2756, 2001; Lonberg, N. and D. Huszar Int. Rev. Immunol 13:65, 1995; and Pollock, et al., J Immunol Methods 231:147, 1999. Methods for making derivatives of antibodies, e.g., humanized, single chain, etc. are known in the art.
  • Immunoassays and flow cytometry sorting techniques such as fluorescence activated cell sorting (FACS) can also be employed to isolate antibodies that are specific for BCMA, CD3, or tumor antigens of interest.
  • FACS fluorescence activated cell sorting
  • Carriers can be active and/or inert.
  • Examples of well-known carriers include polypropylene, polystyrene, polyethylene, dextran, nylon, amylases, glass, natural and modified celluloses, polyacrylamides, agaroses, and magnetite.
  • the nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding antibodies, or will be able to ascertain such, using routine experimentation.
  • the carrier comprises a moiety that targets the myocardium.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors (such as expression vectors disclosed in PCT Publication No. WO 87/04462), which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • expression vectors such as expression vectors disclosed in PCT Publication No. WO 87/04462
  • host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant regions in place of the homologous murine sequences, Morrison et al., Proc. Nat. Acad. Sci. 81:6851, 1984, or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. In that manner, “chimeric” or “hybrid” antibodies are prepared that have the binding specificity of a monoclonal antibody herein.
  • the BCMA or tumor antigen of interest antibodies as described herein can be identified or characterized using methods known in the art, whereby reduction of BCMA or other tumor antigen expression levels are detected and/or measured.
  • a BCMA antibody is identified by incubating a candidate agent with BCMA and monitoring binding and/or attendant reduction of BCMA expression levels.
  • the binding assay may be performed with purified BCMA polypeptide(s), or with cells naturally expressing, or transfected to express, BCMA polypeptide(s).
  • the binding assay is a competitive binding assay, where the ability of a candidate antibody to compete with a known BCMA antibody for BCMA binding is evaluated.
  • the assay may be performed in various formats, including the ELISA format.
  • bioassays known to test the targeted biological activities.
  • bioassays can be used to screen candidates directly.
  • BCMA, CD3, or other tumor antigen antibodies may be characterized using methods well known in the art.
  • one method is to identify the epitope to which it binds, or “epitope mapping.”
  • epitope mapping There are many methods known in the art for mapping and characterizing the location of epitopes on proteins, including solving the crystal structure of an antibody-antigen complex, competition assays, gene fragment expression assays, and synthetic peptide-based assays, as described, for example, in Chapter 11 of Harlow and Lane, Using Antibodies, a Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999.
  • epitope mapping can be used to determine the sequence to which an antibody binds.
  • Epitope mapping is commercially available from various sources, for example, Pepscan Systems (Edelhertweg 15, 8219 PH Lelystad, The Netherlands).
  • the epitope can be a linear epitope, i.e., contained in a single stretch of amino acids, or a conformational epitope formed by a three-dimensional interaction of amino acids that may not necessarily be contained in a single stretch.
  • Peptides of varying lengths e.g., at least 4-6 amino acids long
  • the epitope to which the BCMA, CD3, or other tumor antigen antibody binds can be determined in a systematic screening by using overlapping peptides derived from the BCMA, CD3, or other tumor antigen sequence and determining binding by the BCMA, CD3, or other tumor antigen antibody.
  • the open reading frame encoding BCMA, CD3, or other tumor antigen is fragmented either randomly or by specific genetic constructions and the reactivity of the expressed fragments of BCMA, CD3, or other tumor antigen with the antibody to be tested is determined.
  • the gene fragments may, for example, be produced by PCR and then transcribed and translated into protein in vitro, in the presence of radioactive amino acids.
  • the binding of the antibody to the radioactively labeled BCMA, CD3, or other tumor antigen fragments is then determined by immunoprecipitation and gel electrophoresis.
  • Certain epitopes can also be identified by using large libraries of random peptide sequences displayed on the surface of phage particles (phage libraries). Alternatively, a defined library of overlapping peptide fragments can be tested for binding to the test antibody in simple binding assays.
  • mutagenesis of an antigen binding domain, domain swapping experiments and alanine scanning mutagenesis can be performed to identify residues required, sufficient, and/or necessary for epitope binding.
  • domain swapping experiments can be performed using a mutant BCMA, CD3, or other tumor antigen in which various fragments of the BCMA, CD3, or other tumor antigen protein have been replaced (swapped) with sequences from BCMA from another species (e.g., mouse), or a closely related, but antigenically distinct protein (e.g., Trop-1).
  • a mutant BCMA, CD3, or other tumor antigen in which various fragments of the BCMA, CD3, or other tumor antigen protein have been replaced (swapped) with sequences from BCMA from another species (e.g., mouse), or a closely related, but antigenically distinct protein (e.g., Trop-1).
  • Yet another method which can be used to characterize a BCMA, CD3, or other tumor antigen antibody is to use competition assays with other antibodies known to bind to the same antigen, i.e., various fragments on BCMA, CD3, or other tumor antigen, to determine if the BCMA, CD3, or other tumor antigen antibody binds to the same epitope as other antibodies.
  • Competition assays are well known to those of skill in the art.
  • An expression vector can be used to direct expression of a BCMA, CD3, or other tumor antigen antibody.
  • One skilled in the art is familiar with administration of expression vectors to obtain expression of an exogenous protein in vivo. See, e.g., U.S. Pat. Nos. 6,436,908; 6,413,942; and 6,376,471.
  • Administration of expression vectors includes local or systemic administration, including injection, oral administration, particle gun or catheterized administration, and topical administration.
  • the expression vector is administered directly to the sympathetic trunk or ganglion, or into a coronary artery, atrium, ventrical, or pericardium.
  • Targeted delivery of therapeutic compositions containing an expression vector, or subgenomic polynucleotides can also be used.
  • Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al., Trends Biotechnol., 1993, 11:202; Chiou et al., Gene Therapeutics: Methods And Applications Of Direct Gene Transfer, J. A. Wolff, ed., 1994; Wu et al., J. Biol. Chem., 263:621, 1988; Wu et al., J. Biol. Chem., 269:542, 1994; Zenke et al., Proc. Natl. Acad. Sci. USA, 87:3655, 1990; and Wu et al., J.
  • compositions containing a polynucleotide are administered in a range of about 100 ng to about 200 mg of DNA for local administration in a gene therapy protocol. Concentration ranges of about 500 ng to about 50 mg, about 1 ⁇ g to about 2 mg, about 5 ⁇ g to about 500 ⁇ g, and about 20 ⁇ g to about 100 ⁇ g of DNA can also be used during a gene therapy protocol.
  • the therapeutic polynucleotides and polypeptides can be delivered using gene delivery vehicles.
  • the gene delivery vehicle can be of viral or non-viral origin (see generally, Jolly, Cancer Gene Therapy, 1:51, 1994; Kimura, Human Gene Therapy, 5:845, 1994; Connelly, Human Gene Therapy, 1995, 1:185; and Kaplitt, Nature Genetics, 6:148, 1994). Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence can be either constitutive or regulated.
  • Viral-based vectors for delivery of a desired polynucleotide and expression in a desired cell are well known in the art.
  • Exemplary viral-based vehicles include, but are not limited to, recombinant retroviruses (see, e.g., PCT Publication Nos. WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; WO 93/11230; WO 93/10218; WO 91/02805; U.S. Pat. Nos. 5,219,740 and 4,777,127; GB Pat. No. 2,200,651; and EP Pat. No.
  • alphavirus-based vectors e.g., Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249: ATCC VR-532)
  • AAV adeno-associated virus
  • Non-viral delivery vehicles and methods can also be employed, including, but not limited to, polycationic condensed DNA linked or unlinked to killed adenovirus alone (see, e.g., Curiel, Hum. Gene Ther., 3:147, 1992); ligand-linked DNA (see, e.g., Wu, J. Biol. Chem., 264:16985, 1989); eukaryotic cell delivery vehicles cells (see, e.g., U.S. Pat. No. 5,814,482; PCT Publication Nos. WO 95/07994; WO 96/17072; WO 95/30763; and WO 97/42338) and nucleic charge neutralization or fusion with cell membranes. Naked DNA can also be employed.
  • Exemplary naked DNA introduction methods are described in PCT Publication No. WO 90/11092 and U.S. Pat. No. 5,580,859.
  • Liposomes that can act as gene delivery vehicles are described in U.S. Pat. No. 5,422,120; PCT Publication Nos. WO 95/13796; WO 94/23697; WO 91/14445; and EP 0524968. Additional approaches are described in Philip, Mol. Cell Biol., 14:2411, 1994 and in Woffendin, Proc. Natl. Acad. Sci., 91:1581, 1994.
  • compositions comprising antibodies described herein or made by the methods and having the characteristics described herein.
  • compositions comprise one or more antibodies that bind to CD3 and a tumor antigen (e.g. BCMA), and/or one or more polynucleotides comprising sequences encoding one or more these antibodies.
  • suitable excipients such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • the invention also provides methods of making any of these antibodies.
  • the antibodies of this invention can be made by procedures known in the art.
  • the polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis.
  • Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available.
  • an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Pat. Nos. 5,807,715; 4,816,567; and 6,331,415.
  • Heteroconjugate antibodies comprising two covalently joined antibodies, are also within the scope of the invention. Such antibodies have been used to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (PCT Publication Nos. WO 91/00360 and WO 92/200373; EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents and techniques are well known in the art, and are described in U.S. Pat. No. 4,676,980.
  • Chimeric or hybrid antibodies also may be prepared in vitro using known methods of synthetic protein chemistry, including those involving cross-linking agents.
  • immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond.
  • suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
  • the Fc ⁇ portion can be modified to avoid interaction with Fc ⁇ receptor and the complement and immune systems.
  • the techniques for preparation of such antibodies are described in WO 99/58572.
  • the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. See, for example, U.S. Pat. Nos. 5,997,867 and 5,866,692.
  • the invention encompasses modifications to the antibodies and polypeptides of the invention variants as described herein, including functionally equivalent antibodies which do not significantly affect their properties and variants which have enhanced or decreased activity and/or affinity.
  • the amino acid sequence may be mutated to obtain an antibody with the desired binding affinity to BCMA and/or CD3.
  • Modification of polypeptides is routine practice in the art and need not be described in detail herein.
  • modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or which mature (enhance) the affinity of the polypeptide for its ligand, or use of chemical analogs.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to an epitope tag.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody of an enzyme or a polypeptide which increases the half-life of the antibody in the blood circulation.
  • Substitution variants have at least one amino acid residue in the antibody molecule removed and a different residue inserted in its place.
  • the sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated.
  • Conservative substitutions are shown in Table 5 under the heading of “conservative substitutions.” If such substitutions result in a change in biological activity, then more substantial changes, denominated “exemplary substitutions” in Table 5, or as further described below in reference to amino acid classes, may be introduced and the products screened.
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring amino acid residues are divided into groups based on common side-chain properties:
  • Non-conservative substitutions are made by exchanging a member of one of these classes for another class.
  • cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking.
  • cysteine bond(s) may be added to the antibody to improve its stability, particularly where the antibody is an antibody fragment such as an Fv fragment.
  • Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the variable region. Changes in the variable region can alter binding affinity and/or specificity. In some embodiments, no more than one to five conservative amino acid substitutions are made within a CDR domain. In other embodiments, no more than one to three conservative amino acid substitutions are made within a CDR domain. In still other embodiments, the CDR domain is CDR H3 and/or CDR L3.
  • Modifications also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation.
  • Antibodies are glycosylated at conserved positions in their constant regions (Jefferis and Lund, Chem. Immunol. 65:111-128, 1997; Wright and Morrison, TibTECH 15:26-32, 1997).
  • the oligosaccharide side chains of the immunoglobulins affect the protein's function (Boyd et al., Mol. Immunol. 32:1311-1318, 1996; Wittwe and Howard, Biochem.
  • Oligosaccharides may also serve to target a given glycoprotein to certain molecules based upon specific recognition structures. Glycosylation of antibodies has also been reported to affect antibody-dependent cellular cytotoxicity (ADCC).
  • CHO cells with tetracycline-regulated expression of P(1,4)-N-acetylglucosaminyltransferase III (GnTIII), a glycosyltransferase catalyzing formation of bisecting GIcNAc, was reported to have improved ADCC activity (Umana et al., Mature Biotech. 17:176-180, 1999).
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine, asparagine-X-threonine, and asparagine-X-cysteine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • glycosylation pattern of antibodies may also be altered without altering the underlying nucleotide sequence. Glycosylation largely depends on the host cell used to express the antibody. Since the cell type used for expression of recombinant glycoproteins, e.g. antibodies, as potential therapeutics is rarely the native cell, variations in the glycosylation pattern of the antibodies can be expected (see, e.g. Hse et al., J. Biol. Chem. 272:9062-9070, 1997).
  • factors that affect glycosylation during recombinant production of antibodies include growth mode, media formulation, culture density, oxygenation, pH, purification schemes and the like.
  • Various methods have been proposed to alter the glycosylation pattern achieved in a particular host organism including introducing or overexpressing certain enzymes involved in oligosaccharide production (U.S. Pat. Nos. 5,047,335; 5,510,261 and 5,278,299).
  • Glycosylation or certain types of glycosylation, can be enzymatically removed from the glycoprotein, for example, using endoglycosidase H (Endo H), N-glycosidase F, endoglycosidase F1, endoglycosidase F2, endoglycosidase F3.
  • Endo H endoglycosidase H
  • N-glycosidase F N-glycosidase F
  • endoglycosidase F1 endoglycosidase F2
  • endoglycosidase F3 endoglycosidase F3
  • the recombinant host cell can be genetically engineered to be defective in processing certain types of polysaccharides.
  • Modifications include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay. Modified polypeptides are made using established procedures in the art and can be screened using standard assays known in the art, some of which are described below and in the Examples.
  • the antibody comprises a modified constant region, such as a constant region that has increased affinity to a human Fc gamma receptor, is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate macrophages; or has reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating antibody-dependent cell mediated cytotoxicity (ADCC), or activating microglia.
  • Different modifications of the constant region may be used to achieve optimal level and/or combination of effector functions.
  • the constant region is modified as described in Eur. J. Immunol., 1999, 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 9809951.8.
  • the antibody comprises a human heavy chain IgG2 constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wild type IgG2 sequence). Eur. J. Immunol., 1999, 29:2613-2624.
  • the constant region is aglycosylated for N-linked glycosylation.
  • the constant region is aglycosylated for N-linked glycosylation by mutating the glycosylated amino acid residue or flanking residues that are part of the N-glycosylation recognition sequence in the constant region.
  • N-glycosylation site N297 may be mutated to A, Q, K, or H.
  • the constant region is aglycosylated for N-linked glycosylation.
  • the constant region may be aglycosylated for N-linked glycosylation enzymatically (such as removing carbohydrate by enzyme PNGase), or by expression in a glycosylation deficient host cell.
  • antibody modifications include antibodies that have been modified as described in PCT Publication No. WO 99/58572. These antibodies comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant region of a human immunoglobulin heavy chain. These antibodies are capable of binding the target molecule without triggering significant complement dependent lysis, or cell-mediated destruction of the target. In some embodiments, the effector domain is capable of specifically binding FcRn and/or Fc ⁇ RIIb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain CH 2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy.
  • affinity matured antibodies can be produced by procedures known in the art (Marks et al., Bio/Technology, 10:779-783, 1992; Barbas et al., Proc Nat. Acad. Sci, USA 91:3809-3813, 1994; Schier et al., Gene, 169:147-155, 1995; Yelton et al., J. Immunol., 155:1994-2004, 1995; Jackson et al., J. Immunol., 154(7):3310-9, 1995, Hawkins et al., J. Mol. Biol., 226:889-896, 1992; and PCT Publication No. WO2004/058184).
  • library scanning mutagenesis One way of characterizing a CDR of an antibody and/or altering (such as improving) the binding affinity of a polypeptide, such as an antibody, termed “library scanning mutagenesis”.
  • library scanning mutagenesis works as follows. One or more amino acid positions in the CDR are replaced with two or more (such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) amino acids using art recognized methods. This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of two or more members (if two or more amino acids are substituted at every position).
  • the library also includes a clone comprising the native (unsubstituted) amino acid.
  • a small number of clones, e.g., about 20-80 clones (depending on the complexity of the library), from each library are screened for binding affinity to the target polypeptide (or other binding target), and candidates with increased, the same, decreased, or no binding are identified.
  • Methods for determining binding affinity are well-known in the art. Binding affinity may be determined using BiacoreTM surface plasmon resonance analysis, which detects differences in binding affinity of about 2-fold or greater. BiacoreTM is particularly useful when the starting antibody already binds with a relatively high affinity, for example a K D of about 10 nM or lower. Screening using BiacoreTM surface plasmon resonance is described in the Examples, herein.
  • Binding affinity may be determined using Kinexa Biocensor, scintillation proximity assays, ELISA, ORIGEN immunoassay (IGEN), fluorescence quenching, fluorescence transfer, and/or yeast display. Binding affinity may also be screened using a suitable bioassay.
  • every amino acid position in a CDR is replaced (in some embodiments, one at a time) with all 20 natural amino acids using art recognized mutagenesis methods (some of which are described herein). This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of 20 members (if all 20 amino acids are substituted at every position).
  • the library to be screened comprises substitutions in two or more positions, which may be in the same CDR or in two or more CDRs.
  • the library may comprise substitutions in two or more positions in one CDR.
  • the library may comprise substitution in two or more positions in two or more CDRs.
  • the library may comprise substitution in 3, 4, 5, or more positions, said positions found in two, three, four, five or six CDRs.
  • the substitution may be prepared using low redundancy codons. See, e.g., Table 2 of Balint et al., Gene 137(1):109-18, 1993.
  • the CDR may be CDRH3 and/or CDRL3.
  • the CDR may be one or more of CDRL1, CDRL2, CDRL3, CDRH1, CDRH2, and/or CDRH3.
  • the CDR may be a Kabat CDR, a Chothia CDR, or an extended CDR.
  • Candidates with improved binding may be sequenced, thereby identifying a CDR substitution mutant which results in improved affinity (also termed an “improved” substitution).
  • Candidates that bind may also be sequenced, thereby identifying a CDR substitution which retains binding.
  • candidates each comprising an amino acid substitution at one or more position of one or more CDR
  • candidates with improved binding are also useful for the design of a second library containing at least the original and substituted amino acid at each improved CDR position (i.e., amino acid position in the CDR at which a substitution mutant showed improved binding).
  • Preparation, and screening or selection of this library is discussed further below.
  • Library scanning mutagenesis also provides a means for characterizing a CDR, in so far as the frequency of clones with improved binding, the same binding, decreased binding or no binding also provide information relating to the importance of each amino acid position for the stability of the antibody-antigen complex. For example, if a position of the CDR retains binding when changed to all 20 amino acids, that position is identified as a position that is unlikely to be required for antigen binding. Conversely, if a position of CDR retains binding in only a small percentage of substitutions, that position is identified as a position that is important to CDR function.
  • the library scanning mutagenesis methods generate information regarding positions in the CDRs that can be changed to many different amino acids (including all 20 amino acids), and positions in the CDRs which cannot be changed or which can only be changed to a few amino acids.
  • Candidates with improved affinity may be combined in a second library, which includes the improved amino acid, the original amino acid at that position, and may further include additional substitutions at that position, depending on the complexity of the library that is desired, or permitted using the desired screening or selection method.
  • adjacent amino acid position can be randomized to at least two or more amino acids. Randomization of adjacent amino acids may permit additional conformational flexibility in the mutant CDR, which may in turn, permit or facilitate the introduction of a larger number of improving mutations.
  • the library may also comprise substitution at positions that did not show improved affinity in the first round of screening.
  • the second library is screened or selected for library members with improved and/or altered binding affinity using any method known in the art, including screening using BiacoreTM surface plasmon resonance analysis, and selection using any method known in the art for selection, including phage display, yeast display, and ribosome display.
  • compositions comprising antibodies conjugated (for example, linked) to an agent that facilitate coupling to a solid support (such as biotin or avidin).
  • a solid support such as biotin or avidin.
  • Conjugation generally refers to linking these components as described herein.
  • the linking (which is generally fixing these components in proximate association at least for administration) can be achieved in any number of ways. For example, a direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other.
  • a nucleophilic group such as an amino or sulfhydryl group
  • a carbonyl-containing group such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.
  • the invention provides a method of making any of the polynucleotides described herein.
  • Polynucleotides complementarity to any such sequences are also encompassed by the present invention.
  • Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules.
  • RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence.
  • Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein.
  • Variants preferably exhibit at least about 70% identity, more preferably, at least about 80% identity, yet more preferably, at least about 90% identity, and most preferably, at least about 95% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M. O., 1978, A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D. G.
  • the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof.
  • Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementarity sequence).
  • Suitable “moderately stringent conditions” include prewashing in a solution of 5 ⁇ SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-65° C., 5 ⁇ SSC, overnight; followed by washing twice at 65° C. for 20 minutes with each of 2 ⁇ , 0.5 ⁇ and 0.2 ⁇ SSC containing 0.1% SDS.
  • highly stringent conditions or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5 ⁇ SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 ⁇ Denhardt's solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulf
  • nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
  • a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein.
  • Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome.
  • the polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al., 1989.
  • PCR allows reproduction of DNA sequences.
  • PCR technology is well known in the art and is described in U.S. Pat. Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston, 1994.
  • RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., 1989, supra, for example.
  • Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector.
  • Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, pMB9, CoIE1, pCR1, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
  • Bluescript e.g., pBS SK+
  • shuttle vectors such as pSA3 and pAT28.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462.
  • Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • the vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus).
  • electroporation employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances
  • microprojectile bombardment e.g., where the vector is an infectious agent such as vaccinia virus.
  • infection e.g., where the vector is an infectious agent such as vaccinia virus.
  • the choice of introducing vectors or polynucleotides will often depend on features of the host cell.
  • the invention also provides host cells comprising any of the polynucleotides described herein. Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest.
  • mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462.
  • Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis ) and yeast (such as S. cerevisae, S. pombe ; or K. lactis ).
  • the host cells express the cDNAs at a level of about 5 fold higher, more preferably, 10 fold higher, even more preferably, 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells.
  • Screening the host cells for a specific binding to BCMA or an BCMA domain is effected by an immunoassay or FACS.
  • a cell overexpressing the antibody or protein of interest can be identified.
  • the antibodies e.g., BCMA, CD3, or bispecific
  • the antibody conjugates e.g., BCMA antibody-drug conjugates
  • the present invention are useful in various applications including, but are not limited to, therapeutic treatment methods and diagnostic treatment methods.
  • the invention provides a method for treating a condition associated with BCMA expression in a subject.
  • the method of treating a condition associated with BCMA expression in a subject comprises administering to the subject in need thereof an effective amount of a composition (e.g., pharmaceutical composition) comprising the BCMA antibodies or the BCMA antibody conjugates as described herein.
  • a composition e.g., pharmaceutical composition
  • the conditions associated with BCMA expression include, but are not limited to, abnormal BCMA expression, altered or aberrant BCMA expression, malignant cells expressing BCMA, and a proliferative disorder (e.g., cancer) or autoimmune disorder.
  • the invention provides a method for treating a B-cell related cancer or malignant cells expressing a tumor antigen.
  • a method of treating a B-cell related cancer in a subject in need thereof comprising a) providing the bispecific antibody as described herein, and b) administering said bispecific antibody to said patient.
  • a method of treating a condition associated with malignant cells expressing a tumor antigen in a subject comprising administering to the subject in need thereof an effective amount of a pharmaceutical composition comprising the bispecific antibody as described herein.
  • a method of treating a cancer in a subject comprising administering to the subject in need thereof an effective amount of a composition comprising the antibodies (e.g., BCMA, or CD3-BCMA bispecific antibodies) or the BCMA antibody conjugates as described herein.
  • a composition comprising the antibodies (e.g., BCMA, or CD3-BCMA bispecific antibodies) or the BCMA antibody conjugates as described herein.
  • cancer can be a B-cell related cancer including, but are not limited to, multiple myeloma, malignant plasma cell neoplasm, Hodgkin's lymphoma, nodular lymphocyte predominant Hodgkin's lymphoma, Kahler's disease and Myelomatosis, plasma cell leukemia, plasmacytoma, B-cell prolymphocytic leukemia, hairy cell leukemia, B-cell non-Hodgkin's lymphoma (NHL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), follicular lymphoma, Burkitt's lymphoma, marginal zone lymphoma, mantle cell lymphoma, large cell lymphoma, precursor B-lymphoblastic lymphoma, myeloid leukemia, Waldenstrom's macroglobulienemia, diffuse
  • provided is a method of inhibiting tumor growth or progression in a subject who has malignant cells expressing BCMA comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein.
  • a method of inhibiting metastasis cells expressing BCMA in a subject comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein.
  • provided is a method of inducing tumor regression in malignant cells in a subject comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein.
  • provided is a method of treating an autoimmune disorder in a subject comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein.
  • autoimmune disorders include, but are not limited to, systemic lupus erythematosus, rheumatoid arthritis, diabetes (Type 1), multiple sclerosis, Addison's disease, celiac disease, dermatomyositis, Graves' disease, hashimoto's thyroiditis, hashimoto's encephalopathy, Myasthenia gravis, pernicious anemia, reactive arthritis, Sjogren syndrome, acute disseminated encephalomyelitis, agammaglobulinemia, amyotrophic lateral sclerosis, ankylosing spondylitis, antiphospholipid syndrome, antisynthetase syndrome, atopic allergy, atopic dermatitis, autoimmune enteropathy, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune lymphoproliferative syndrome, autoimmune peripheral neuropathy, autoimmune pancreatitis, autoimmune polyendorcrine syndrome, autoimmune peripheral neuropathy
  • the invention provides an effective amount of a composition (e.g., pharmaceutical composition) comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression in a subject in need thereof.
  • a condition e.g., cancer or autoimmune disorder
  • a composition e.g., pharmaceutical composition
  • the invention provides the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for use in treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression in a subject in need thereof.
  • a condition e.g., cancer or autoimmune disorder
  • provided is the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inhibiting tumor growth or progression in a subject who has malignant cells expressing BCMA.
  • provided is the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inhibiting metastasis of malignant cells expressing BCMA in a subject in need thereof.
  • provided provided is the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inducing tumor regression in a subject who has malignant cells expressing BCMA.
  • the invention provides a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression.
  • a condition e.g., cancer or autoimmune disorder
  • provided is a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting tumor growth or progression.
  • provided is a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting metastasis of malignant cells expressing BCMA.
  • provided is a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inducing tumor regression.
  • the antibodies e.g., BCMA or CD3-BCMA bispecific
  • a detectable moiety such as an imaging agent and an enzyme-substrate label.
  • the antibodies as described herein can also be used for in vivo diagnostic assays, such as in vivo imaging (e.g., PET or SPECT), or a staining reagent.
  • the methods described herein further comprise a step of treating a subject with an additional form of therapy.
  • the additional form of therapy is an additional anti-cancer therapy including, but not limited to, chemotherapy, radiation, surgery, hormone therapy, and/or additional immunotherapy.
  • the additional form of therapy comprises administering one or more therapeutic agent in addition to the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein.
  • the one or more therapeutic agent can be a chemotherapeutic agents including, but not limited to, a second antibody (e.g., an anti-VEGF (Vascular Endothelial Growth Factor) antibody (e.g., AVASTIN®), an anti-HER2 antibody (e.g., HERCEPTIN®), an anti-CD25 antibody, an anti-CD33 antibody, an anti-CD20 antibody (e.g., RITUXAN®), an anti-mucin-like glycoprotein antibody, an anti-TNF antibody, and/or an epidermal growth factor receptor (EGFR) antibody (e.g., ERBITUX®)), an angiogenesis inhibitor, a cytotoxic agent (e.g., anthracyclines (e.g., daunorubicin, dox
  • a method of treating multiple myeloma comprising administering to a patient need thereof an effective amount of a composition comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein and one or more other therapeutic agent such as a chemotherapeutic agent (e.g., doxorubicin or carfilzomib) or thalidomide or its derivative thereof (e.g., lenalidomide (REVLIMID®)).
  • chemotherapeutic agent e.g., doxorubicin or carfilzomib
  • thalidomide or its derivative thereof e.g., lenalidomide (REVLIMID®
  • the one or more other therapeutic agent is selecting from the group consisting of bortezomib (e.g., VELCADE®), melphalan, prednisone, doxorubicin, lenalidomide, thalidomide, prednisone, carmustine, etoposide, cisplatin, cyclophosphamide, carfilzomib, and vincristine.
  • the other therapeutic agent is bortezomib (e.g., VELCADE®), melphalan, lenalidomide (REVLIMID®), carfilzomib, doxorubicin, or prednisone.
  • a method of treating multiple myeloma comprising administering to a patient need thereof an effective amount of a composition comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein and one or more other therapeutic agent selecting from the group consisting of bortezomib, lenalidomide, carfilzomib, and doxorubicin.
  • the patient is relapsing or refractory to previous multiple myeloma therapy.
  • the antibodies e.g., BCMA or CD3-BCMA bispecific
  • the BCMA antibody conjugates can be administered to an individual via any suitable route. It should be understood by persons skilled in the art that the examples described herein are not intended to be limiting but to be illustrative of the techniques available.
  • the antibody e.g., BCMA or CD3-BCMA bispecific
  • the BCMA antibody conjugate is administered to an individual in accord with known methods, such as intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, intracranial, transdermal, subcutaneous, intra-articular, sublingually, intrasynovial, via insufflation, intrathecal, oral, inhalation or topical routes.
  • Administration can be systemic, e.g., intravenous administration, or localized.
  • nebulizers for liquid formulations, including jet nebulizers and ultrasonic nebulizers are useful for administration.
  • Liquid formulations can be directly nebulized and lyophilized powder can be nebulized after reconstitution.
  • the antibody e.g., BCMA or CD3-BCMA bispecific
  • the BCMA antibody conjugate can be aerosolized using a fluorocarbon formulation and a metered dose inhaler, or inhaled as a lyophilized and milled powder.
  • the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate is administered via site-specific or targeted local delivery techniques.
  • site-specific or targeted local delivery techniques include various implantable depot sources of the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate or local delivery catheters, such as infusion catheters, indwelling catheters, or needle catheters, synthetic grafts, adventitial wraps, shunts and stents or other implantable devices, site specific carriers, direct injection, or direct application. See, e.g., PCT Publication No. WO 00/53211 and U.S. Pat. No. 5,981,568.
  • the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate may be used for administration.
  • the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate may be administered neat.
  • the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate and a pharmaceutically acceptable excipient may be in various formulations.
  • Pharmaceutically acceptable excipients are known in the art, and are relatively inert substances that facilitate administration of a pharmacologically effective substance. For example, an excipient can give form or consistency, or act as a diluent.
  • Suitable excipients include but are not limited to stabilizing agents, wetting and emulsifying agents, salts for varying osmolarity, encapsulating agents, buffers, and skin penetration enhancers. Excipients as well as formulations for parenteral and nonparenteral drug delivery are set forth in Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005.
  • these agents are formulated for administration by injection (e.g., intraperitoneally, intravenously, subcutaneously, intramuscularly, etc.). Accordingly, these agents can be combined with pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like.
  • pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like.
  • the particular dosage regimen, i.e., dose, timing and repetition, will depend on the particular individual and that individual's medical history.
  • the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein can be administered using any suitable method, including by injection (e.g., intraperitoneally, intravenously, subcutaneously, intramuscularly, etc.).
  • the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate can also be administered via inhalation, as described herein.
  • an initial candidate dosage can be about 2 mg/kg.
  • a typical daily dosage might range from about any of 3 pg/kg to 30 pg/kg to 300 pg/kg to 3 mg/kg, to 30 mg/kg, to 100 mg/kg or more, depending on the factors mentioned above. For example, dosage of about 1 mg/kg, about 2.5 mg/kg, about 5 mg/kg, about 10 mg/kg, and about 25 mg/kg may be used.
  • the treatment is sustained until a desired suppression of symptoms occurs or until sufficient therapeutic levels are achieved, for example, to inhibit or delay tumor growth/progression or metatstasis of cancer cells.
  • An exemplary dosing regimen comprises administering an initial dose of about 2 mg/kg, followed by a weekly maintenance dose of about 1 mg/kg of the antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate, or followed by a maintenance dose of about 1 mg/kg every other week.
  • Other exemplary dosing regimen comprises administering increasing doses (e.g., initial dose of 1 mg/kg and gradual increase to one or more higher doses every week or longer time period).
  • Other dosage regimens may also be useful, depending on the pattern of pharmacokinetic decay that the practitioner wishes to achieve. For example, in some embodiments, dosing from one to four times a week is contemplated.
  • dosing once a month or once every other month or every three months is contemplated.
  • the progress of this therapy is easily monitored by conventional techniques and assays.
  • the dosing regimen (including the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate used) can vary over time.
  • an antibody e.g., BCMA or CD3-BCMA bispecific
  • a BCMA antibody conjugate will depend on the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate (or compositions thereof) employed, the type and severity of symptoms to be treated, whether the agent is administered for therapeutic purposes, previous therapy, the patient's clinical history and response to the agent, the patient's clearance rate for the administered agent, and the discretion of the attending physician.
  • the clinician will administer an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate until a dosage is reached that achieves the desired result. Dose and/or frequency can vary over course of treatment.
  • Empirical considerations such as the half-life, generally will contribute to the determination of the dosage.
  • antibodies that are compatible with the human immune system such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's immune system.
  • Frequency of administration may be determined and adjusted over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of symptoms, e.g., tumor growth inhibition or delay, etc.
  • sustained continuous release formulations of antibodies e.g., BCMA or CD3-BCMA bispecific
  • BCMA antibody conjugates may be appropriate.
  • Various formulations and devices for achieving sustained release are known in the art.
  • dosages for an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate may be determined empirically in individuals who have been given one or more administration(s) of the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate. Individuals are given incremental dosages of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate. To assess efficacy, an indicator of the disease can be followed.
  • an antibody e.g., BCMA or CD3-BCMA bispecific
  • an BCMA antibody conjugate in accordance with the method in the present invention can be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate may be essentially continuous over a preselected period of time or may be in a series of spaced doses.
  • more than one antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate may be present. At least one, at least two, at least three, at least four, at least five different or more antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate can be present. Generally, those antibodies (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugates may have complementary activities that do not adversely affect each other. For example, one or more of the following antibody may be used: a first BCMA or CD3 antibody directed to one epitope on BCMA or CD3 and a second BCMA or CD3 antibody directed to a different epitope on BCMA or CD3.
  • Therapeutic formulations of the antibody e.g., BCMA or CD3-BCMA bispecific
  • the BCMA antibody conjugate used in accordance with the present invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and may comprise buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine,
  • Liposomes containing the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate are prepared by methods known in the art, such as described in Epstein, et al., Proc. Natl. Acad. Sci. USA 82:3688, 1985; Hwang, et al., Proc. Natl Acad. Sci. USA 77:4030, 1980; and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • PEG-PE PEG-derivatized phosphatidylethanolamine
  • the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or ‘poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and 7 ethyl-L-glutamate copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-( ⁇ )-3-hydroxybutyric acid.
  • LUPRON DEPOTTM injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate
  • sucrose acetate isobutyrate sucrose acetate isobutyrate
  • poly-D-( ⁇ )-3-hydroxybutyric acid poly-D-( ⁇ )-3-hydroxybutyric acid.
  • compositions to be used for in vivo administration must be sterile. This is readily accomplished by, for example, filtration through sterile filtration membranes.
  • Therapeutic antibody e.g., BCMA or CD3-BCMA bispecific
  • BCMA antibody conjugate compositions are generally placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • compositions according to the present invention may be in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, or administration by inhalation or insufflation.
  • the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof.
  • a pharmaceutical carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water
  • a pharmaceutical carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • Suitable surface-active agents include, in particular, non-ionic agents, such as polyoxyethylenesorbitans (e.g. TweenTM 20, 40, 60, 80 or 85) and other sorbitans (e.g. SpanTM 20, 40, 60, 80 or 85).
  • Compositions with a surface-active agent will conveniently comprise between 0.05 and 5% surface-active agent, and can be between 0.1 and 2.5%. It will be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
  • Suitable emulsions may be prepared using commercially available fat emulsions, such as IntralipidTM, LiposynTM, InfonutrolTM, LipofundinTM and LipiphysanTM.
  • the active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g. soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g. egg phospholipids, soybean phospholipids or soybean lecithin) and water.
  • an oil e.g. soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil
  • a phospholipid e.g. egg phospholipids, soybean phospholipids or soybean lecithin
  • other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion.
  • Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%.
  • the fat emulsion can comprise fat droplets between 0.1 and 1.0 ⁇ m, particularly 0.1 and 0.5 ⁇ m, and have a pH in the range of 5.5 to 8.0.
  • the emulsion compositions can be those prepared by mixing an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water).
  • an antibody e.g., BCMA or CD3-BCMA bispecific
  • a BCMA antibody conjugate with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water).
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulised by use of gases. Nebulised solutions may be breathed directly from the nebulising device or the nebulising device may be attached to a face mask, tent or intermittent positive pressure breathing machine.
  • Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • compositions used in the methods of the invention comprise an effective amount of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate as described herein. Examples of such compositions, as well as how to formulate, are also described in an earlier section and below.
  • the composition comprises one or more antibodies (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugates.
  • BCMA antibody or CD3-BCMA bispecific antibody recognizes human BCMA or CD3-BCMA.
  • the BCMA or CD3-BCMA antibody is a human antibody, a humanized antibody, or a chimeric antibody.
  • the BCMA antibody or CD3-BCMA antibody comprises a constant region that is capable of triggering a desired immune response, such as antibody-mediated lysis or ADCC. In other embodiments, the BCMA antibody or CD3-BCMA antibody comprises a constant region that does not trigger an unwanted or undesirable immune response, such as antibody-mediated lysis or ADCC.
  • compositions can comprise more than one antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate (e.g., a mixture of BCMA antibodies or CD3-BCMA bispecific antibodies that recognize different epitopes of BCMA or CD3 and BCMA).
  • BCMA antibody conjugate e.g., a mixture of BCMA antibodies or CD3-BCMA bispecific antibodies that recognize different epitopes of BCMA or CD3 and BCMA.
  • Other exemplary compositions comprise more than one BCMA antibody, CD3-BCMA antibody, or BCMA antibody conjugate that recognize the same epitope(s), or different species of BCMA antibodies, CD3-BCMA bispecific antibodies, or BCMA antibody conjugate that bind to different epitopes of BCMA (e.g., human BCMA) or CD3 and BCMA (human CD3 and BCMA).
  • composition used in the present invention can further comprise pharmaceutically acceptable carriers, excipients, or stabilizers (Remington: The Science and practice of Pharmacy 21st Ed., 2005, Lippincott Williams and Wilkins, Ed. K. E. Hoover), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • Kits of the invention include one or more containers comprising the BCMA antibody, CD3-BCMA bispecific antibody, or the BCMA antibody conjugate as described herein and instructions for use in accordance with any of the methods of the invention described herein.
  • these instructions comprise a description of administration of the BCMA antibody, CD3-BCMA bispecific antibody, or the BCMA antibody conjugate for the above described therapeutic treatments.
  • the instructions relating to the use of the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
  • the containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses.
  • Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
  • kits of this invention are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like.
  • packages for use in combination with a specific device such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump.
  • a kit may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container may also have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a BCMA antibody, CD3-BCMA bispecific antibody, or a BCMA antibody conjugate.
  • the container may further comprise a second pharmaceutically active agent.
  • Kits may optionally provide additional components such as buffers and interpretive information.
  • the kit comprises a container and a label or package insert(s) on or associated with the container.
  • Example 1 Determination of Kinetics and Affinity of hBCMA/Human IgG Interactions at 25° C. and/or 37° C.
  • This example determines the kinetics and affinity of various anti-BCMA antibodies at 25° C. and 37° C.
  • Channels were activated in the analyte (horizontal) direction by injecting a mixture of 1 mM ECD and 0.25 mM NHS for 3 minutes at a flow rate of 30 ⁇ L/min.
  • IgGs were immobilized on the activated spots by injecting them in the ligand (vertical) direction at 20 pg/mL in 10 mM Acetate pH 4.5 buffer for 1.5 minutes at 30 pg/mL.
  • the activated surfaces were blocked by injecting 1M ethanolamine, pH 8.5 in the analyte direction for 3 minutes at 30 pL/min.
  • the analysis temperature for the hBCMA binding analysis was 37° C. or 25° C. in a running buffer of HBS-T+, supplemented with 1 mg/mL BSA.
  • a kinetic titration method was employed for the interaction analysis as described in Abdiche, et al.
  • the hBCMA (human BCMA) analyte was injected in the analyte direction using a series of injections from low to high concentration.
  • the concentrations used were 0.08 nM, 0.4 nM, 2 nM, 10 nM and 50 nM (a 5-membered series, with a 5-fold dilution factor and top concentration of 50 nM).
  • the association time for a given analyte dilution was two minutes.
  • the sensorgrams were double-referenced and fit to a 1:1 Langmuir with mass transport kinetic titration model in BIAevaluation Software version 4.1.1 (GE Lifesciences, Piscataway, NJ). The sensorgrams and fits are shown in FIG. 1 , and the kinetics and affinity parameters for various anti-BCMA antibodies of the present invention are shown in Tables 6A-6C.
  • This example demonstrates binding of BCMA positive tumor cells by various BCMA antibodies of the present invention.
  • Binding of human anti-hBCMA expressed in mouse IgG2a were assessed on BCMA-expressing cells (KMS12BM, L363, MM1S and KMS12PE) by flow cytometry. 250,000 cells were incubated with 0.5 ug antibody in 100 uL binding buffer (PBS (Phosphate Buffered Saline)+0.2% BSA (Bovine Serum Albumin)), followed by incubation with Alex Fluor 647 conjugated anti-mouse IgG (Biolegend).
  • PBS Phosphate Buffered Saline
  • BSA Bovine Serum Albumin
  • Table 7 shows MFI (mean fluorescence intensity) on BCMA positive tumor cells by various BCMA antibodies (e.g., Combo_Rd4_0.6 nM_C29, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C16, and P6E01/H3TAQ)
  • BCMA antibodies e.g., Combo_Rd4_0.6 nM_C29, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C16, and P6E01/H3TAQ
  • This example illustrates the efficacy of the anti-BCMA ADCs in BCMA positive cells.
  • Human anti-BCMA (L3.PY/P6E01, L3.PY/H3.TAQ, Combo_Rd4_0.6 nM_C29, A02_Rd4_6 nM_C01, and A02_Rd4_6 nM C16) antibodies were expressed as human IgG1 subtypes engineered with glutamine-containing transglutaminase (“Q”) tags (e.g. LCQ05, H7c, N297A, N297Q, N297A/H7c, N297Q/LCQ05) for drug antibody ratios (DAR) of 2, 4, and 6.
  • Q glutamine-containing transglutaminase
  • the transglutaminase tags can be engineered at the light chain, heavy chain, or a combination of light and heavy chains.
  • the transglutaminase tag (e.g., Q) is engineered at site of the antibody, such as at position 297 of the human IgG (EU numbering scheme).
  • the wild-type amino acid asparagine (N) is substituted with glutamine or alanine at position 297 of the BCMA antibody (N297Q or N297A) of the present invention.
  • Anti-BCMA antibody conjugation to Aur0101, Aur3377, and Aur0131 was then achieved via microbial transglutaminase-catalyzed transamidation reaction between the anti-BCMA antibody carrying a targeted glutamine or glutamine tag at the specific site (e.g., carboxyl terminus or amino terminus of the heavy chain or light chain, position 297, or at another site of the antibody) and an amine-containing derivative of the payload (e.g., MMAD, Aur0101, Aur3377, or Aur0131).
  • a targeted glutamine or glutamine tag at the specific site (e.g., carboxyl terminus or amino terminus of the heavy chain or light chain, position 297, or at another site of the antibody) and an amine-containing derivative of the payload (e.g., MMAD, Aur0101, Aur3377, or Aur0131).
  • the wild-type amino acid lysine at position 222, 340, or 370 was replaced with amino acid arginine (“K222R”, “K340R”, or “K370R”).
  • K222R amino acid arginine
  • the K222R substitution was found to have the surprising effect of resulting in more homogenous antibody and payload conjugate, better intermolecular crosslinking between the antibody and the payload, and/or significant decrease in interchain crosslinking with the glutamine tag on the C-terminus of the antibody light chain.
  • the glutamine on the antibody acted as an acyl donor, and the amine-containing compound acted as an acyl acceptor (amine donor).
  • Purified anti-BCMA antibody in the concentration of 1-150 ⁇ M was incubated with a 5-100 molar excess acyl acceptor, ranging between 5 ⁇ M-15 mM, in the presence of 0.23-0.55% (w/v) Streptoverticillium mobaraense transglutaminase (ACTIVATM, Ajinomoto, Japan) in 10-1000 mM NaCl, and 25 mM MES, HEPES [4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid] or Tris HCl buffer at pH range 6.2-8.8.
  • the reaction conditions were adjusted for individual acyl acceptor derivatives, and the optimal efficiency and specificity were typically observed for 33 ⁇ M antibody, 0.67 mM derivative, and 0.378% (w/v) transglutaminase in 75 mM NaCl, 25 mM Tris HCl, pH 8.5.
  • the antibody was purified on Butyl Sepharose High Performance (Butyl HP) resin (GE Healthcare, Waukesha, WI) using standard chromatography methods known to persons skilled in the art, such as commercial hydrophobic interaction chromatography from GE Healthcare.
  • Target expressing (MM1.S, KMS12BM and L363) cells were then seeded on clear bottom plates at 3000 cells/well. Cells were treated with 4-fold serially diluted antibody-drug conjugates in triplicates. Cell viability was determined by CellTiter-Glo® Luminescent Cell Viability Assay 96 (Promega, Madison WI) 96 hours after treatment. Relative cell viability was determined as percentage of untreated control. EC50 was calculated by Prism software. Table 8 shows that all human anti-BCMA antibodies of the present invention conjugated to cytotoxic agent 0101, 3377, and 0131 through transglutaminase tags and linkers exert potent cell killing activity in BCMA expressing cells.
  • This example illustrates the in vivo efficacy of the anti-BCMA ADCs in the MM1 S orthotopic multiple myeloma model.
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA).
  • IVIS Spectrum CT Perkin Elmer, MA
  • Living Image 4.4 Caliper Life Sciences, Alameda, CA.
  • the animals were randomized into groups and a single dose of a human anti-BCMA antibody conjugated with 1) LCQ05/K222R-vc0101 at the C-terminus of the antibody light chain and control conjugates were administered through bolus tail vein injection. Animals were terminated when they exhibit hindlimb paralysis, an endpoint for MM1.S orthotopic models.
  • NNC negative control
  • This example also illustrates the in vivo efficacy of the anti-BCMA ADCs in the MM1.S orthotopic multiple myeloma models.
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA).
  • FIG. 3 shows that a single dose of human anti-BCMA L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-0131 and 2) H7c/N297A/K222R-amino-PEG6-C2-3377 resulted in tumor regression.
  • This example also illustrates the in vivo efficacy of the anti-BCMA ADCs in the KMS12BM orthotopic multiple myeloma models
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin are captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA).
  • FIG. 4 shows that a single dose of human anti-BCMA L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-Aur0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101 resulted in tumor inhibition.
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA).
  • FIG. 5 shows that a single dose of human anti-BCMA COMBO_Rd4_0.6 nM_C29 antibody conjugated with groups 1)-4) above resulted in tumor regression starting at 0.1 mg/kg and tumor inhibition up to 100 days starting at 0.75 mg/kg.
  • This example describes the generation and purification of the heterodimeric antibodies of the present application.
  • variable region of the human specific anti-CD3 antibody was cloned into a human IgG1 or IgG2AA containing the following mutations 221R, 228R, and K409R; or 223R, 225R, 228R, and K409R, respectively, and referred as hIgG1 RRR or IgG2AA-RRRR.
  • variable region of the anti-target antibody was cloned into a human IgG1 or IgG2AA containing the following mutations 221E, 228E, L368E or 223E, 225E, 228E and L368E, respectively, and referred as hIgG1EEE or hIgG2AA-EEEE.
  • Heterodimers were prepared by incubation of the anti-CD3 IgG1 or IgG2AA having hIgG1 RRR or IgG2AA-RRRR mutations with an anti-target antibody having hIgG1 EEE or hIgG2AA-EEEE mutations in PBS with 1 mM or 2 mM GSH for 24 hrs at 37° C. as described in International Patent Application No. PCT/US2011/036419 (WO2011/143545).
  • the heterodimer was purified by ion exchange chromatography, as described below.
  • heterodimers were purified by ion exchange chromatography. Briefly, analytical ion exchange separation of the Fc-hetero and Fc-homodimers was carried out on Agilent 1100 quaternary pump LC system (Agilent Inc, Santa Clara, CA, USA) equipped with weak cation exchange DIONEX Propac WCX-10G (4 ⁇ 50 mm) column. Proteins were injected in 5% buffer A (20 mM MES pH 5.4) and eluted in a gradient from 25% to 75% buffer B (20 mM MES pH 5.4 and 500 mM NaCl) over a 20 minute period with 1 ml/min flow rate.
  • Agilent 1100 quaternary pump LC system Agilent 1100 quaternary pump LC system (Agilent Inc, Santa Clara, CA, USA) equipped with weak cation exchange DIONEX Propac WCX-10G (4 ⁇ 50 mm) column. Proteins were injected in 5% buffer A (20 mM MES pH
  • Example 9 Determination of Kinetics and Affinity of hCD3/Human IgG Interactions at 25° C. and/or 37° C.
  • This example determines the kinetics and affinity of various anti-CD3 antibodies at 25° C. and 37° C.
  • Channels were activated in the analyte (horizontal) direction by injecting a mixture of 1 mM ECD and 0.25 mM NHS for 3 minutes at a flow rate of 30 ⁇ L/min.
  • IgGs were immobilized on the activated spots by injecting them in the ligand (vertical) direction at 20 pg/mL in 10 mM Acetate pH 4.5 buffer for 1.5 minutes at 30 ⁇ g/mL.
  • the activated surfaces were blocked by injecting 1M ethanolamine, pH 8.5 in the analyte direction for 3 minutes at 30 ⁇ L/min.
  • the analysis temperature for the hCD3 binding analysis was 37° C. or 25° C. in a running buffer of HBS-T+, supplemented with 1 mg/mL BSA.
  • a kinetic titration method was employed for the interaction analysis as described in Abdiche, et al.
  • the hCD3 (human CD3) analyte was injected in the analyte direction using a series of injections from low to high concentration.
  • the concentrations used were 0.08 nM, 0.4 nM, 2 nM, 10 nM and 50 nM (a 5-membered series, with a 5-fold dilution factor and top concentration of 50 nM).
  • the association time for a given analyte dilution was two minutes.
  • This example demonstrates the efficacy of the anti-CD3-anti-CD20 bispecific antibodies in CD20+ cells.
  • Efficacy was determined by measuring B cells and T cells in peripheral blood by flow cytometry. Whole blood was collected at the time points indicated and kept at 4° C. until analysis. Erythrocytes were lysed with ACK buffer (Gibco) for 5 minutes at room temperature and white blood cells were pelleted by centrifugation. Cells were stained for 1 hr. at 4° C. with a cocktail containing fluorescently labeled antibodies recognizing cyno CD19 (Beckman Coulter), CD45, CD4, CD8, Ki67 (BD Biosciences) in PBS+2% FBS.
  • Ki67 analysis cells were first stained with CD4 and CD8, then fix/permeabilized with BD cyotfix/cytoperm kit (BD Biosciences) according to manufacturer's instructions prior to intracellular staining for Ki67. Acquisition of cells on a BD LSRII flow cytometer was carried out immediately after staining.
  • the resulting B cell count was graphed as a percentage of the pre-study B cell count in FIGS. 6 A- 6 F .
  • Prolonged B cell depletion following a single dose was achieved with doses as low as 2 ug/kg. B cell depletion was seen at all doses. The duration of the depletion effect was dose dependent.
  • the resulting CD8+ T cell count was graphed as percentage of the pre-study CD8+ T cell count in FIGS. 7 A- 7 F . After an initial relocalization, T cell levels were restored to baseline levels or above for the duration of the study.
  • This example demonstrates the efficacy of the monovalent anti-CD3 antibody on T cell kinetics and activation.
  • the resulting T cell count was graphed as percentage of the pre-study CD8+ T cell count in FIGS. 8 A and 8 B .
  • Ki67+ T cells increased and peaked between day 3 and day 7 post dose, indicating T cell activation.
  • NNC/h2B4 there was no increase in Ki67+ T cells.
  • This example demonstrates the effect of anti-CD3 arm affinity on B cell depletion.
  • FIGS. 9 A- 9 D the resulting B cell count was graphed as a percentage of the pre-study B cell count. Efficacy of B cell depletion correlates to anti-CD3 arm affinity.
  • This example illustrates the in vitro cytotoxicity of the Anti-BCMA/CD3 hIgG2AA Bispecific in BCMA Positive Cells.
  • Human anti-BCMA (P5A2, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C16, P5C1, C01_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C22, Combo_Rd4_0.6 nM_C29, L3PY/H3TAQ and A02_Rd4_6 nM_C01) and human anti-CD3 (H2B4) antibodies were expressed as human IgG2dA engineered with EEEE for bispecific exchange as described in Example 8.
  • CD3+ T cells from PBMC were negatively selected using Pan T Cell Isolation kit, human (Miltenyi, San Diego CA).
  • Target expressing (KMS12PE, L363 and Molp8) cells and CD3+ T-cells were seeded on clear U-bottom plates at 20000 and 100000 cells/well respectively.
  • Cells were treated with 10-fold serially diluted bispecific antibody in triplicates.
  • Cell death was determined by CytoTox 96 ⁇ Non-Radioactive Cytotoxicity Assay (Promega, Madison WI) 20 hours after treatment. Cell cytotoxicity was determined as percentage of untreated effector plus target control wells. EC50 was calculated by Prism software. Table 10 shows that all human anti-BCMA_H2B4 bispecific antibodies exert cell killing activity in BCMA expressing cells.
  • This example illustrates the in vitro T cell activation/proliferation of the anti-CD3 cloned from mouse hybridoma in human/cynomolgus PBMC cells for antibody screening.
  • Human anti-CD3 antibodies were cloned from immunized mouse, expressed as mouse IgG1, and purified by Protein A affinity beads.
  • Human/Cynomolgus peripheral blood mononuclear cells (hu/cyPBMC) were prepared by Ficoll (Density: 1.083 g/mL, GE) density gradient centrifugation from blood filters obtained from local blood banks. Erythrocytes were removed by incubating in LCK buffer (155 mM NH4Cl, 10 mM KHCO3, 100 mM EDTA; Gibco) for 3 minutes at room temperature. Cells were centrifuged for 5 min at 600 g.
  • LCK buffer 155 mM NH4Cl, 10 mM KHCO3, 100 mM EDTA; Gibco
  • the supernatant containing the lysed erythrocytes was discarded, and the PBMC were washed twice in 50 ml 1 ⁇ PBS/1% BSA/1 mM EDTA.
  • the pelleted cells were adjusted to 10 7 cells per ml in culture media, X-VIVO-15, serum free media (Lonza,), and the PBMC were seeded as 10 6 (100ul) per-well to round bottom 96 well tissue culture plates.
  • Selected Abs are 10 ⁇ serial diluted from 1000ng to 1ng per mL for mixing with human PBMC and 5 ⁇ serial diluted from 5000ng to 200ng per mL for mixing with cynomolgus PBMC.
  • PBMC T cell proliferation by 3 H-thymidine incorporation 2 day cultures were performed in triplicate. During the final 16 h of culture 3 H-thymidine (0.5 mCi/well) was added, and incorporation was measured. Cells are harvested and lysed, DNA is captured onto glass-fiber filter. Radioactivity (cpm) as measure for proliferation by counting on a scintillation beta-counter.
  • FIGS. 10 A and 10 B show that the selected anti-CD3 1A4, 1C10, 2B4, and 7A3 antibodies had Thymidine incorporation reading on human and cynomolgus PBMC (peripheral blood mononuclear cells).
  • Table 11 shows their KDs by Biacore measurement.
  • In vitro characterization shows anti-CD3 1C10 and 2B4 antibodies are similar to the positive control SP34 anti-CD3 antibody (BD Biosciences)
  • This example illustrates the in vitro cytotoxicity of the anti-EpCam/CD3 Bispecific in SW480 mixed with healthy donor isolated Pan T cells.
  • A Anti-CD antibodies h2B4-1d, TK, hnpsTK, and yaesTK
  • Human anti-CD3 (h2B4-1d (or h2B4), h2B4-TK (or h2B4-VH-wt VL_TK), h2B4-hnpsTK (or h2B4-VH-hnps VL_TK), and h2B4-yaesTK (or h2B4-VH-yaes VL_TK)) antibodies and human anti-EpCam antibodies were expressed as human IgG2dA engineered with RRRR or EEEE for bispecific exchange as describe in Example 8.
  • the SW480 was selected as target cell line for cell killing assay and the effector cells and human T cells were purified from human peripheral blood mononuclear cells (huPBMC).
  • Target and effector cells were seeded in 96-well, round bottom plates in cell culture medium containing 5% fetal bovine serum (FBS). The number of target cells was kept constant at 2 ⁇ 10 4 cells/well.
  • FBS fetal bovine serum
  • LDH lactate dehydrogenase
  • FIGS. 11 A and 11 B show that all human anti-EpCam_h2B4 bispecific antibodies had cell killing activity on in vitro setting, and antibody mediated T cell activation was monitoring by T cell activation marker.
  • Table 12A shows their EC50.
  • Table 12B shows Biacore KD on the bispecific antibody format.
  • Human anti-CD3 h2B4(h2B4_1d) and h25A8 (m25A8, h25A8-B12, and h25A8-B13) and human anti-EpCam antibodies were expressed as human IgG2dA engineered with RRRR or EEEE for bispecific exchange as describe in Example 8.
  • FIGS. 11 C and 11 D show that all human anti-EpCam_anti-CD3 bispecific antibodies had cell killing activity on in vitro setting, and antibody mediated T cell activation was monitoring by T cell activation marker.
  • Table 12C shows the EC50 of in vitro cell killing.
  • Table 12D shows the Biacore kinetics on the bispecific antibody format at 37° C.
  • Table 12E shows in vitro characterization using SEC-MALS (Size Exclusion Chromatography with Multi-Angle Light Scattering) and DSC (Differential Scanning Calorimety).
  • This example illustrates the in vitro cytotoxicity of the anti-BCMA/CD3 Bispecific in primary myeloma cells.
  • Human anti-BCMA (P5A2, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C16, Combo_Rd4_0.6 nM_029, and P6E01 L3PY/H3TAQ) and human anti-CD3 (h2B4) antibodies were expressed as human IgG2dA engineered with EEEE or RRRR for bispecific exchange as describe in Example 8.
  • Total bone marrow mononuclear cells from myeloma patients were seeded in clear U-bottom plates at total bone marrow mononuclear cell numbers that resulted in 3000-5000 myeloma cells/well.
  • Cells were treated with 10-fold serially diluted bispecific antibody. Five days after treatment, total viable cells were determined by flow cytometry using antibodies to CD138 and CD38 (Biolegend, CA). Cells were incubated with antibodies at 4° in PBS+0.5% FIBS for 30 minutes. Cells were washed and Fixable Viability Dye eFluor 780 (eBioscience, Inc., CA) in PBS was added to the cells for 30 minutes at 4°.
  • Table 13A shows that all human anti-BCMA_h2B4 bispecific antibodies have cell killing activity on myeloma patient samples and patient T cells are functional effector cells.
  • Table 13B shows killing of one anti-BCMA bispecific on multiple myeloma patient samples with different effector to target (E:T) ratio.
  • Example 17 ELISPOT of Antibody Secreting Cells from Cynomolgus Monkeys Administered with Anti-BCMA/CD3 Bispecific Antibodies
  • This example illustrates the depletion of IgG secreting cells in cynomolgus monkey with anti-BCMA/CD3 bispecific antibodies.
  • PBMC Peripheral blood mononuclear cells
  • a sample of bone marrow (from femur) was collected by flushing with approximately 5 mL of 100% fetal bovine serum (FBS) and then single cell suspensions were prepared by suspending flushed marrow in 50 ml of buffer.
  • FBS fetal bovine serum
  • PBMC or bone marrow cells were added to triplicate wells at specified concentrations (PBMCs at 5 ⁇ 10 5 /well, and bone marrow cells at 2 ⁇ 10 5 /well), and then cells were serially diluted in the plate. After an overnight incubation, plates were washed and a biotinylated detection antibody was added.
  • IgG spots were visualized using TMB substrate solution and counted using the ImmunoSpot Imaging Analyzer system (CTL) and ImmunoSpot 5.1 software. Data were expressed as the mean (+/ ⁇ SD) number of IgG-secreting cells from triplicate samples.
  • IgG-secreting cell count in PBMC and in bone marrow are listed in Tables 14A and 14B, respectively.
  • Depletion of IgG-secreting cells was seen for both anti-BCMA/CD3 bispecific antibodies in PBMC as compared to pre-dose and bone marrow as compared to vehicle and negative control. A dose dependent effect was seen in the bone marrow.
  • Example 18 Anti-BCMA/CD3 Bispecific Induce Tumor Regression and Inhibition in MM1.S Tumor Model
  • This example illustrates tumor regression and inhibition in an orthotopic MM1.S myeloma model.
  • mice were irradiated with 100cGy using RS 2000 Biological Research Irradiator (RAD Source Technolgies, GA). Five million MM1.S LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female Nod/Scid/IL2Rg ⁇ / ⁇ (NSG) animals.
  • D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enabled monitoring of tumor burden.
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 15E6 for all animals, the animals were injected through bolus tail vein with 20 million expanded T cells from PBMC.
  • pan-T cells purchased from AllCells (Alameda, CA) were activated with human T Cell Activation/Expansion Kit (Miltenyi, San Diego, CA). After three days, 15U/mL of IL2 (ebioscience, San Diego, CA) was added every two days until day 11. Cells were harvested, activation/expansion beads were magnetically removed, and cells were washed and resuspended in PBS.
  • mice were imaged as described above and animals were randomized into groups of seven mice; A02_Rd4_0.6 nM_C01 at 0.03 mg/kg and 0.3 mg/kg and Combo_Rd4_0.6 nM_C29 at 0.03 mg/kg, 0.1 mg/kg and 0.3 mg/kg.
  • a single dose of human anti-BCMA/CD3 (h2B4-VH-wt VL_TK) bispecific and negative (NNC) control bispecific antibody was administered through bolus tail vein injection. Animals were sacrificed when they exhibited hindlimb paralysis, an endpoint for MM1.S orthotopic model.
  • FIG. 12 shows that a single dose of human anti-BCMA/CD3 bispecific antibody resulted in tumor regression in a dose-dependent manner.
  • This example illustrates tumor regression with two doses of anti-BCMA/CD3 bispecific antibodies in an orthotopic Molp8 myeloma model.
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA).
  • IVIS Spectrum CT Perkin Elmer, MA
  • Living Image 4.4 Caliper Life Sciences, Alameda, CA.
  • mice Two days post T cell injection, mice were dosed with bispecific antibodies. Animals were sacrificed when they exhibited weight loss of more than 15%, an endpoint for Molp8 orthotopic model.
  • FIG. 13 shows that two doses of human anti-BCMA/CD3 (h2B4-VH-wt VL_TK) bispecific antibody resulted in increased tumor regression.
  • This example demonstrates no opposing effects on anti-BCMA/CD3 bispecific antibodies when combined with bortezomib or lenalidomide and better potency with anti-BCMA/CD3 bispecific antibodies as compared to bortezomib and lenalidomie combined.
  • Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). The animals were injected through bolus tail vein on day 7 with 20 million expanded T cells as described in Example 18.
  • mice Two days post T cell injection, mice were imaged and randomized into five groups of seven mice with an average of 17E6 total flux/group: 1) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg, 2) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg and 1 mg/kg bortezomib, 3) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg and 50 mg/kg lenalidomide, 4) 1 mg/kg bortezomib and 50 mg/kg lenalidomide and 5) vehicle.
  • Anti-BCMA/CD3 (in PBS) was injected through bolus tail vein injection, bortezomib (in PBS) was administrated via intraperitoneal injection and lenalidomide (30% PEG400/5% propylene glycol/0.5% Tween80) via oral gavage.
  • Vehicle consisted of 30% PEG400/5% propylene glycol/0.5% Tween80, which was administrated via oral gavage. Animals were sacrificed when they exhibit weight loss of more than 15%, an endpoint for Molp8 orthotopic model.
  • Example 21 In Vitro Study of Anti-BCMA/CD3 Bispecific in Combination with Lenalidomide, Carfilzomib or Doxorubicin on OPM2 Cell Line
  • This example illustrates no adverse effects on T cell function when combined with carfilzomib, doxorubicin, and lenalidomide for anti-BCMA/CD3 bispecific antibody activity as compared to the bispecific antibody alone.
  • CD3+ T cells from PBMC were negatively selected using Pan T Cell Isolation kit, human (Miltenyi, San Diego CA). OPM2 cells and CD3+ T-cells were seeded in clear U-bottom plates 20000 and 100000 cells/well, respectively. OPM2 and CD3+ T cells were first incubated with the standard of care for two hours at 37°. 1.56 nM carfilzomib and 6.25 nM doxorubicin were diluted in PBS containing 0.02% DMSO. Lenalidomide was diluted in PBS containing 0.1% DMSO at 195 nM. Cells were treated with 10-fold serially diluted bispecific antibody.
  • This example illustrates synergistic effects on anti-BCMA/CD3 bispecific function when combined with carfilzomib and lenalidomide as compared to each molecule alone.
  • CD3+ T cells from PBMC were negatively selected using Pan T Cell Isolation kit, human (Miltenyi, San Diego CA). KMS12BM cells and CD3+ T-cells were seeded in clear U-bottom plates 20000 and 100000 cells/well, respectively. Cells were treated with 0.017 nM anti-BCMA/CD3 bispecific in combination with carfilzomib and a range of concentration for lenalidomide. 1.25 nM carfilzomib was diluted in PBS containing 0.02% DMSO. Lenalidomide was diluted in PBS containing 0.1% DMSO starting at 4 uM diluted 4-fold.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Endocrinology (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to antibodies, e.g., full length antibodies or antigen binding fragments thereof, that specifically bind to BCMA (B-Cell Maturation Antigen) and/or CD3 (Cluster of Differentiation 3). The invention also relates to antibody conjugates (e.g., antibody-drug-conjugates) comprising the BCMA antibodies, compositions comprising the BCMA antibodies, and methods of using the BCMA antibodies and their conjugates for treating conditions associated with cells expressing BCMA (e.g., cancer or autoimmune disease). The invention further relates to heteromultimeric antibodies that specifically bind to CD3 and a tumor cell antigen, (e.g., bispecific antibodies that specifically bind to CD3 and BCMA). Compositions comprising such heteromultimeric antibodies, methods for producing and purifying such heterodimeric antibodies, and their use in diagnostics and therapeutics are also provided.

Description

    RELATED APPLICATIONS
  • This application is a continuation application of U.S. patent application Ser. No. 17/005,073, filed Aug. 27, 2020, which is a divisional of U.S. patent application Ser. No. 16/017,974, filed Jun. 25, 2018, now U.S. Pat. No. 10,793,635, which is a divisional of Ser. No. 15/878,344, filed Jan. 23, 2018, now U.S. Pat. No. 10,040,860, which is a divisional of U.S. application Ser. No. 15/085,644, filed on Mar. 30, 2016, now U.S. Pat. No. 9,969,809, which claims the benefits of U.S. Provisional Application No. 62/146,843 filed Apr. 13, 2015, U.S. Provisional Application No. 62/146,504 filed Apr. 13, 2015, and U.S. Provisional Application No. 62/301,582 filed Feb. 29, 2016, all of which are hereby incorporated by reference in their entireties.
  • REFERENCE TO SEQUENCE LISTING
  • This application is being filed electronically via Patent Center and includes an electronically submitted sequence listing in .xml format. The .xml file contains a sequence listing entitled “PC72209F_SEQListing_ST26.xml” created on Aug. 23, 2023 and having a size of 669 KB. The sequence listing contained in this .xml file is part of the specification and is incorporated herein by reference in its entirety.
  • FIELD
  • The present invention relates to antibodies, e.g., full length antibodies or antigen binding fragments thereof, that specifically bind to BCMA (B-Cell Maturation Antigen) and/or CD3 (Cluster of Differentiation 3). The invention also relates to antibody conjugates (e.g., antibody-drug-conjugates) comprising the BCMA antibodies, compositions comprising the BCMA antibodies, and methods of using the BCMA antibodies and their conjugates for treating conditions associated with cells expressing BCMA (e.g., cancer or autoimmune disease). The invention further relates to heteromultimeric antibodies that specifically bind to CD3 and a tumor cell antigen, (e.g., bispecific antibodies that specifically bind to CD3 and BCMA). Compositions comprising such heteromultimeric antibodies, methods for producing and purifying such heterodimeric antibodies, and their use in diagnostics and therapeutics are also provided.
  • BACKGROUND
  • B-cell maturation antigen (BCMA, CD269, or TNFRSF17) is a member of the tumor necrosis factor receptor (TNFR) superfamily. BCMA was identified in a malignant human T cell lymphoma containing a t(4;16) translocation. The gene is selectively expressed in the B-cell lineage with the highest expression in plasma blasts and plasma cells, antibody secreting cells. BCMA binds two ligands, B-cell activation factor (BAFF) (also called B-lymphocyte stimulator (BLyS) and APOL-related leukocyte expressed ligand (TALL-1)) and a proliferation-inducing ligand (APRIL) with affinity of 1 uM and 16 nM, respectively. Binding of APRIL or BAFF to BCMA promotes a signaling cascade involving NF-kappa B, Elk-1, c-Jun N-terminal kinase and the p38 mitogen-activated protein kinase, which produce signals for cell survival and proliferation.
  • BCMA is also expressed on malignant B cells and several cancers that involve B lymphocytes including multiple myeloma, plasmacytoma, Hodgkin's Lymphoma, and chronic lymphocytic leukemia. In autoimmune diseases where plasmablasts are involved such as systemic lupus erythematosus (SLE) and rheumatoid arthritis, BCMA expressing antibody-producing cells secrete autoantibodies that attack self.
  • In the case of multiple myeloma, about 24,000 new cases are newly diagnosed in the United States each year, and this number represents about 15% of the newly diagnosed hematological cancers in the United States. An average of 11,000 deaths result from multiple myeloma each year, and the average 5-year survival rate is about 44%, with median survival of 50-55 months. Current treatment for multiple myeloma is focused on plasma cells apoptosis and/or decreasing osteoclast activity (e.g., chemotherapy, thalidomide, lenalidomide, bisphosphonates, and/or proteasome inhibitors such as bortezomib (VELCADE®) or carfilzomib). However, multiple myeloma remains an incurable disease, and almost all patients have developed resistance to these agents and eventually relapse. Accordingly, an alternative treatment to multiple myeloma, such as using an anti-BCMA antagonist including antibodies and other immunotherapeutic agents (e.g. bispecific antibodies or antibody-drug conjugates), would make a superior therapeutic agent.
  • SUMMARY
  • The invention disclosed herein is directed to therapeutic antibodies that bind to BCMA and/or CD3. Antibody conjugates (e.g., antibody-drug conjugates) comprising BCMA are also provided. Further, the heteromultimeric antibodies (e.g., bispecific antibodies) that specifically bind to CD3 and a tumor cell antigen (e.g., bispecific antibodies that specifically bind to CD3 and BCMA) are also provided.
  • In one aspect, the invention provides an isolated antibody, or an antigen binding fragment thereof, which specifically binds to B-Cell Maturation Antigen (BCMA), wherein the antibody comprises (a) a heavy chain variable (VH) region comprising (i) a VH complementary determining region one (CDR1) comprising the sequence SYX1MX2, wherein X1 is A or P; and X2 is T, N, or S (SEQ ID NO: 301), GFTFX1SY, wherein X1 is G or S (SEQ ID NO: 302), or GFTFX1SYX2MX3, wherein X1 is G or S, X2 is A or P; and X3 is T, N, or S (SEQ ID NO: 303); (ii) a VH CDR2 comprising the sequence AX1X2X3X4GX5X6X7X8YADX9X10KG, wherein X1 is I, V, T, H, L, A, or C; X2 is S, D, G, T, I, L, F, M, or V; X3 is G, Y, L, H, D, A, S, or M; X4 is S, Q, T, A, F, or W; X5 is G or T; X6 is N, S, P, Y, W, or F; X7 is S, T, I, L, T, A, R, V, K, G, or C; X8 is F, Y, P, W, H, or G; X9 is V, R, or L; and X10 is G or T (SEQ ID NO: 305), or X1X2X3X4X5X6, wherein X1 is S, V, I, D, G, T, L, F, or M; X2 is G, Y, L, H, D, A, S, or M; X3 is S, G, F, or W; X4 is G or S; X5 is G or T; and X6 is N, S, P, Y, or W (SEQ ID NO: 306); and iii) a VH CDR3 comprising the sequence VSPIX1X2X3X4, wherein X1 is A or Y; X2 is A or S; and X3 is G, Q, L, P, or E (SEQ ID NO: 307), or YWPMX1X2, wherein X1 is D, S, T, or A; and X2 is I, S, L, P, or D (SEQ ID NO: 308); and/or (b) a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence X1X2X3X4X5X6X7X8X9X10X11X12, wherein X1 is R, G, W, A, or C; X2 is A, P, G, L, C, or S; X3 is S, G, or R; X4 is Q, C, E, V, or I; X5 is S, P, G, A, R, or D; X6 is V, G, I, or L; X7 is S, E, D, P, or G; X8 is S, P, F, A, M, E, V, N, D, or Y; X9 is I, T, V, E, S, A, M, Q, Y, H, R, or F; X10 is Y or F; X11 is L, W, or P; and X12 is A, S, or G (SEQ ID NO: 309); (ii) a VL CDR2 comprising the sequence X1ASX2RAX3, wherein X1 is G or D; X2 is S or I; and X3 is T or P (SEQ ID NO: 310); and (iii) a VL CDR3 comprising the sequence QQYX1X2X3PX4T, wherein X1 is G, Q, E, L, F, A, S, M, K, R, or Y; X2 is S, R, T, G, V, F, Y, D, A, H, V, E, K, or C; X3 is W, F, or S; and X4 is L or I (SEQ ID NO: 311), or QQYX1X2X3PX4, wherein X1 is G, Q, E, L, F, A, S, M, R, K, or Y; X2 is S, R, T, G, R, V, D, A, H, E, K, C, F, or Y; X3 is W, S, or F; and X4 is L or I (SEQ ID NO: 312).
  • In another aspect, the invention provides an isolated antibody, or an antigen binding fragment thereof, which specifically binds to BCMA, wherein the antibody comprises: a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 125, 127, 313, 314, 363, or 365; and/or a VL region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38, 40, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 317, 81, 82, 84, 85, 86, 88, 89, 90, 91, 93, 94, 96, 98, 100, 102, 103, 105, 107, 108, 109, 111, 113, 115, 116, 117, 119, 121, 123, 124, 126, 128, 315, 316, or 364. In some embodiments, the VH region comprises (i) a VH CDR1 comprising SEQ ID NO:150, 151, 152, 156, or 157; (ii) a VH CDR2 comprising SEQ ID NO: 169, 154, 194, 159, 195, 196, 162, 158, 198, 177, 178, 199, 200, 201, 202, 203, 204, 206, 207, 208, 172, 203, or 204; and (iii) a VH CDR3 comprising SEQ ID NO: 155, 161, 197, 205, or 164; and/or wherein the VL region comprises (i) a VL CDR1 comprising SEQ ID NO: 209, 271, 273, 275, 251, 277, 260, 279, 245, 283, 285, 287, 290, 292, 235, 297, or 299; (ii) a VL CDR2 comprising SEQ ID NO: 221; and (iii) a VL CDR3 comprising SEQ ID NO: 225, 272, 274, 276, 278, 280, 281, 282, 284, 286, 288, 289, 291, 293, 294, 229, 296, 298, or 300. In some embodiments, the VH region comprises the sequence shown in SEQ ID NO: 112 or a variant with one or several conservative amino acid substitutions in residues that are not within a CDR and/or the VL region comprises the amino acid sequence shown in SEQ ID NO: 38 or a variant thereof with one or several amino acid substitutions in amino acids that are not within a CDR. In some embodiments, the antibody comprises a light chain comprising the sequence shown in SEQ ID NO: 357 and a heavy chain comprising the sequence shown in SEQ ID NO: 358. In some embodiments, the antibody comprises a VH region produced by the expression vector with ATCC Accession No. PTA-122094. In some embodiments, the antibody comprises a VL region produced by the expression vector with ATCC Accession No. PTA-122093.
  • In another aspect, the invention provides an isolated antibody comprising an acyl donor glutamine-containing tag engineered at a specific site of the BCMA antibody of the present invention. In some embodiments, the tag comprises an amino acid sequence selected from the group consisting of Q, LQG, LLQGG (SEQ ID NO:318), LLQG (SEQ ID NO:454), LSLSQG (SEQ ID NO: 455), GGGLLQGG (SEQ ID NO: 456), GLLQG (SEQ ID NO: 457), LLQ, GSPLAQSHGG (SEQ ID NO: 458), GLLQGGG (SEQ ID NO: 459), GLLQGG (SEQ ID NO: 460), GLLQ (SEQ ID NO: 461), LLQLLQGA (SEQ ID NO: 462), LLQGA (SEQ ID NO: 463), LLQYQGA (SEQ ID NO: 464), LLQGSG (SEQ ID NO: 465), LLQYQG (SEQ ID NO: 466), LLQLLQG (SEQ ID NO: 467), SLLQG (SEQ ID NO: 468), LLQLQ (SEQ ID NO: 469), LLQLLQ (SEQ ID NO: 470), LLQGR (SEQ ID NO: 471), LLQGPP (SEQ ID NO: 472), LLQGPA (SEQ ID NO: 473), GGLLQGPP (SEQ ID NO: 474), GGLLQGA (SEQ ID NO: 475), LLQGPGK (SEQ ID NO: 476), LLQGPG (SEQ ID NO: 477), LLQGP (SEQ ID NO: 478), LLQP (SEQ ID NO: 479), LLQPGK (SEQ ID NO: 480), LLQAPGK (SEQ ID NO: 481), LLQGAPG (SEQ ID NO: 482), LLQGAP (SEQ ID NO: 483), and LLQLQG (SEQ ID NO: 484).
  • In one variation, the invention provides an isolated antibody comprising an acyl donor glutamine-containing tag and an amino acid modification at position 222, 340, or 370 of the BCMA antibody of the present invention. In some embodiments, the amino acid modification is a substitution from lysine to arginine.
  • In some embodiments, the BCMA antibody of the present invention further comprises a linker. In some embodiments, the linker is selected from the group consisting of Ac-Lys-Gly (acetyl-lysine-glycine), aminocaproic acid, Ac-Lys-p-Ala (acetyl-lysine-β-alanine), amino-PEG2 (polyethylene glycol)-C2, amino-PEG3-C2, amino-PEG6-C2, Ac-Lys-Val-Cit-PABC (acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl), amino-PEG6-C2-Val-Cit-PABC, aminocaproyl-Val-Cit-PABC, [(3R,5R)-1-{3-[2-(2-aminoethoxy)ethoxy]propanoyl}piperidine-3,5-diyl]bis-Val-Cit-PABC, [(3S,5S)-1-{3-[2-(2-aminoethoxy)ethoxy]propanoyl}piperidine-3,5-diyl]bis-Val-Cit-PABC, putrescine, and Ac-Lys-putrescine.
  • In another aspect, the invention provides a conjugate of the BCMA antibody or the antigen binding fragment as described herein, wherein the antibody or the antigen binding fragment is conjugated to an agent, wherein the agent is selected from the group consisting of a cytotoxic agent, an immunomodulating agent, an imaging agent, a therapeutic protein, a biopolymer, and an oligonucleotide. In some embodiments, the agent is a cytotoxic agent including, but not limited to, an anthracycline, an auristatin, a camptothecin, a combretastatin, a dolastatin, a duocarmycin, an enediyne, a geldanamycin, an indolino-benzodiazepine dimer, a maytansine, a puromycin, a pyrrolobenzodiazepine dimer, a taxane, a vinca alkaloid, a tubulysin, a hemiasterlin, a spliceostatin, a pladienolide, and stereoisomers, isosteres, analogs, or derivatives thereof. For example, the cytotoxic agent is MMAD (Monomethyl Auristatin D), 0101 (2-methylalanyl-N-[(3R,4S,5S)-3-methoxy-1-{(2S)-2-[(1R,2R)-1-methoxy-2-methyl-3-oxo-3-{[(1 S)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino}propyl]pyrrolidin-1-yl}-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), 3377 (N,2-dimethylalanyl-N-{(1 S,2R)-4-{(2S)-2-[(1R,2R)-3-{[(1 S)-1-carboxyl-2-phenylethyl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-2-methoxy-1-[(1 S)-1-methylpropyl]-4-oxobutyl}-N-methyl-L-valinamide), 0131 (2-methyl-L-proly-N-[(3R,4S,5S)-1-{(2S)-2-[(1R,2R)-3-{[(1S)-1-carboxy-2-phenylethyl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), or 0121(2-methyl-L-proly-N-[(3R,4S,5S)-1-{(2S)-2-[(1R,2R)-3-{[(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide).
  • In some embodiments, the present invention provides a conjugate comprising the formula: antibody-(acyl donor glutamine-containing tag)-(linker)-(cytotoxic agent). In some embodiments, the acyl donor glutamine-containing tag comprises an amino acid sequence LLQG (SEQ ID NO: 319) and/or GGLLQGPP (SEQ ID NO: 339) and wherein the linker comprises acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl or amino-PEG6-C2. In some embodiments, the conjugate is selected from the group consisting of 1) antibody-GGLLQGPP (SEQ ID NO: 339)-(acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl (AcLys-VC-PABC))-0101; 2) antibody-LLQG (SEQ ID NO: 319)-amino-PEG6-C2-0131; and 3) antibody-LLQG (SEQ ID NO: 319)-amino-PEG6-C2-3377. In some embodiments, the conjugate further comprises an amino acid substitution from lysine to arginine at antibody position 222. In some embodiments, the conjugate further comprises amino acid substitutions at antibody position N297Q or N297A.
  • In another aspect, provided is a method of producing the BCMA antibody as described herein, comprising culturing the host cell under conditions that result in production of the BCMA antibody, and isolating the BCMA antibody from the host cell or culture.
  • In another aspect, the invention provides a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression. In some embodiments, provided is a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting tumor growth or progression. In some embodiments, provided is a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting metastasis of malignant cells expressing BCMA. In some embodiments, provided is a use of the BCMA antibodies or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inducing tumor regression.
  • In another aspect, the invention provides an isolated antibody, or an antigen binding fragment thereof, which specifically binds to CD3, wherein the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443,445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399. In some embodiments, the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387. In some embodiments, the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 340, 343, 430, 431, 435, or 440, 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341, 433, 452, or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342, 432, 434, 437, 438, 439, 446, or 453. In some embodiments, the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 407, or 408 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336, 404, 405, or 417; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335 or 406; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343 or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341 or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342 or 439. In some embodiments, the antibody comprises a VH region produced by the expression vector with ATCC Accession No. PTA-122513. In some embodiments, the antibody comprises a VL region produced by the expression vector with ATCC Accession No. PTA-122512.
  • In another aspect, provided is an isolated antibody which specifically binds to CD3 and competes with the anti-CD3 antibody of the present invention as described herein.
  • In another aspect, the invention provides a bispecific antibody wherein the bispecific antibody is a full-length human antibody, comprising a first antibody variable domain of the bispecific antibody capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen located on the human immune effector cell, and comprising a second antibody variable domain of the bispecific antibody capable of specifically binding to a target antigen, wherein the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443, 445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399. In some embodiments, the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH CDR1-comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 340, 343, 430, 431, 435, or 440, 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341, 433, 452, or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342, 432, 434, 437, 438, 439, 446, or 453. In some embodiments, the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387; and the second antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 112; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 38.
  • In some embodiments, the second antibody variable domain comprises (a) a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence SYX1MX2, wherein X1 is A or P; and X2 is T, N, or S (SEQ ID NO: 301), GFTFX1SY, wherein X1 is G or S (SEQ ID NO: 302), or GFTFX1SYX2MX3, wherein X1 is G or S, X2 is A or P; and X3 is T, N, or S (SEQ ID NO: 303); (ii) a VH CDR2 comprising the sequence AX1X2X3X4GX5X6X7X8YADX9X10KG, wherein X1 is I, V, T, H, L, A, or C; X2 is S, D, G, T, I, L, F, M, or V; X3 is G, Y, L, H, D, A, S, or M; X4 is S, Q, T, A, F, or W; X5 is G or T; X6 is N, S, P, Y, W, or F; X7 is S, T, I, L, T, A, R, V, K, G, or C; X8 is F, Y, P, W, H, or G; X9 is V, R, or L; and X10 is G or T (SEQ ID NO: 305), or X1X2X3X4X5X6, wherein X1 is S, V, I, D, G, T, L, F, or M; X2 is G, Y, L, H, D, A, S, or M; X3 is S, G, F, or W; X4 is G or S; X5 is G or T; and X6 is N, S, P, Y, or W (SEQ ID NO: 306); and iii) a VH CDR3 comprising the sequence VSPIX1X2X3X4 wherein X1 is A or Y; X2 is A or S; and X3 is G, Q, L, P, or E (SEQ ID NO: 307), or YWPMX1X2, wherein X1 is D, S, T, or A; and X2 is I, S, L, P, or D (SEQ ID NO: 308); and/or (b) a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence X1X2X3X4X5X6X7X8X9X10X11X12, wherein X1 is R, G, W, A, or C; X2 is A, P, G, L, C, or S; X3 is S, G, or R; X4 is Q, C, E, V, or I; X5 is S, L, P, G, A, R, or D; X6 is V, G, or I; X7 is S, E, D, or P; X8 is S, P, F, A, M, E, V, N, D, or Y; X9 is I, T, V, E, S, A, M, Q, Y, H, or R; X10 is Y or F; X11 is L, W, or P; and X12 is A, S, or G (SEQ ID NO: 309); (ii) a VL CDR2 comprising the sequence X1ASX2RAX3, wherein X1 is G or D; X2 is S or I; and X3 is T or P (SEQ ID NO: 310); and (iii) a VL CDR3 comprising the sequence QQYX1X2X3PX4T, wherein X1 is G, Q, E, L, F, A, S, M, K, R, or Y; X2 is S, R, T, G, V, F, Y, D, A, H, V, E, K, or C; X3 is W, F, or S; and X4 is L or I (SEQ ID NO: 311), or QQYX1X2X3PX4, wherein X1 is G, Q, E, L, F, A, S, M, R, K, or Y; X2 is S, R, T, G, R, V, D, A, H, E, K, C, F, or Y; X3 is W, S, or F; and X4 is L or I (SEQ ID NO: 312). In some embodiments, the second antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO:150, 151, 152, 156, 157, 348, 349, 353, 354, or 355; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 169, 154, 194, 159, 195, 196, 162, 158, 198, 177, 178, 199, 200, 201, 202, 203, 204, 206, 207, 208, 172, 203, 204, 350, 351, 356 or 357; and (iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 155, 161, 197, 205, 164, or 352, or 358; and/or wherein the light chain variable (VL) region comprises (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209, 271, 273, 275, 251, 277, 260, 279, 245, 283, 285, 287, 290, 292, 235, 297, 299, or 361; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221, 359 or 362; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 211, 225, 272, 274, 276, 278, 280, 281, 282, 284, 286, 288, 289, 291, 293, 294, 229, 296, 298, 300 or 360.
  • In some embodiments, (a) the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 407, or 408(ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336, 417, 404, or 405; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335 or 406; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343 or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341 or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342 or 439; and (b) the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 151, 156, or 157; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 158 or 159; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and/or wherein the light chain variable (VL) region comprises (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 225.
  • In some embodiments, both the first and the second antibody variable domains of the bispecific antibody comprise amino acid modifications at positions 223, 225, and 228 in the hinge region and at position 409 or 368 (EU numbering scheme) in the CH3 region of a human IgG2 (SEQ ID NO: 493). In some embodiments, the bispecific antibody as described herein further comprises an amino acid modification at position 265 of the human IgG2.
  • In another aspect, the invention provides pharmaceutical compositions comprising any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • In another aspect, the invention also provides cell lines that recombinantly produce any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • In another aspect, the invention also provides nucleic acids encoding any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein. The invention also provides nucleic acids encoding a heavy chain variable region and/or a light chain variable region of any of these antibodies described herein.
  • The invention also provides kits comprising an effective amount of any of the antibodies (e.g., BCMA, CD3, or bispecific) or the conjugates thereof (e.g., BCMA antibody-drug conjugate) described herein.
  • The invention also provides methods of treating a condition (e.g., tumor growth/progression inhibition; metastasis of malignant cells expressing BCMA inhibition; tumor regression induction in subjects with malignant cells expressing BCMA) in subjects in need thereof comprising providing the isolated antibodies (e.g., BCMA) or binding fragments, bispecific antibodies (BCMA-CD3 bispecifics), or the conjugates thereof (e.g., BCMA antibody-drug conjugates) thereof described herein and administering said antibodies or conjugates to said subject.
  • Also provided are methods of treating a condition associated with malignant cells expressing a tumor antigen in a subject comprising administering to a subject in need thereof an effective amount of the pharmaceutical compositions of the invention. In some embodiments, the condition is cancer. In some embodiments, the cancer is a B-cell related cancer selecting from the group consisting of multiple myeloma, malignant plasma cell neoplasm, Hodgkin's lymphoma, nodular lymphocyte predominant Hodgkin's lymphoma, Kahler's disease and Myelomatosis, plasma cell leukemia, plasmacytoma, B-cell prolymphocytic leukemia, hairy cell leukemia, B-cell non-Hodgkin's lymphoma (NHL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), follicular lymphoma, Burkitt's lymphoma, marginal zone lymphoma, mantle cell lymphoma, large cell lymphoma, precursor B-lymphoblastic lymphoma, myeloid leukemia, Waldenstrom's macroglobulienemia, diffuse large B cell lymphoma, follicular lymphoma, marginal zone lymphoma, mucosa-associated lymphatic tissue lymphoma, small cell lymphocytic lymphoma, mantle cell lymphoma, Burkitt lymphoma, primary mediastinal (thymic) large B-cell lymphoma, lymphoplasmactyic lymphoma, Waldenström macroglobulinemia, nodal marginal zone B cell lymphoma, splenic marginal zone lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, lymphomatoid granulomatosis, T cell/histiocyte-rich large B-cell lymphoma, primary central nervous system lymphoma, primary cutaneous diffuse large B-cell lymphoma (leg type), EBV positive diffuse large B-cell lymphoma of the elderly, diffuse large B-cell lymphoma associated with inflammation, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, large B-cell lymphoma arising in HHV8-associated multicentric Castleman disease, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma, and other B-cell related lymphoma. In some embodiments, the condition is an autoimmune disorder, such as systemic lupus erythematosus or rheumatoid arthritis.
  • In some embodiments, the antibodies described herein comprise a constant region. In some embodiments, the antibodies described herein are of the human IgG1, IgG2 or IgG2Aa, IgG3, or IgG4 subclass. In some embodiments, the antibodies described herein comprise a glycosylated constant region. In some embodiments, the antibodies described herein comprise a constant region having increased binding affinity to one or more human Fc gamma receptor(s).
  • BRIEF DESCRIPTION OF THE FIGURES/DRAWINGS
  • FIG. 1A-FIG. 1D depict the double-referenced sensorgrams with fit curves for interactions between selected anti-BCMA antibodies of the present invention and human BCMA.
  • FIG. 2 depicts in vivo efficacy studies of various anti-BCMA ADCs in the MM1 S orthotopic multiple myeloma model, including P6E01_VHVL-AcLys-Val-Cit-PABC-Aur0101; P5A2_VHVL-AcLys-Val-Cit-PABC-Aur0101; P5C1_VHVL-AcLys-Val-Cit-PABC-Aur0101; P4G4-AcLys-Val-Cit-PABC-Aur0101; and P1A11-AcLys-Val-Cit-PABC-Aur0101. NNC is a negative control non-BCMA antibody. “LCQ05” and “LCQ04” correspond to glutamine-containing transglutaminase tag SEQ ID NOs: 474 and 475, respectively.
  • FIG. 3 depicts in vivo efficacy of the anti-BCMA ADCs in the MM1S orthotopic multiple myeloma model, including L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101. NNC is a control non-BCMA antibody. “LCQ05” and H7c correspond to glutamine-containing transglutaminase tag SEQ ID NO: 474 and SEQ ID NO: 454, respectively
  • FIG. 4 also depicts in vivo efficacy of the anti-BCMA ADCs in the MM1S orthotopic multiple myeloma model, including L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-Aur0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101. NNC is a control non-BCMA antibody (antibody-N297Q/K222R-AcLys-VC-PABC-0101). “LCQ05” and H7c correspond to glutamine-containing transglutaminase tag SEQ ID NO: 474 and SEQ ID NO: 454, respectively.
  • FIG. 5 also depicts in vivo efficacy of an anti-BCMA ADC in the MM1S orthotopic multiple myeloma model. Anti-BCMA antibody COMBO_Rd4_0.6 nM-C29 (“Combo C29 DI) is conjugated to H7c/N297A/K222R-amino-PEG6-C2-131 at doses ranging from 0.1 mg/kg, 0.38 mg/kg, 0.75 mg/kg, 1.5 mg/kg in comparison to NNC, a control non-BCMA antibody (antibody-N297Q/K222R-AcLys-VC-PABC-0101) at 3 mg/kg. H7c correspond to glutamine-containing transglutaminase tag SEQ ID NO: 454.
  • FIG. 6A-FIG. 6F depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. B cell depletion following a single dose of bispecific antibody is shown as a percentage of prestudy counts.
  • FIG. 7A-FIG. 7F depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. CD8+ T cell kinetics were tracked following a single dose of bispecific antibody.
  • FIG. 8A and FIG. 8B depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. The effect of the monovalent CD3 antibody on T cell kinetics and proliferation was analyzed.
  • FIGS. 9A-FIG. 9D depict the in vivo efficacy of an anti-CD3/anti-CD20 bispecific antibody in cynomolgus monkeys. The effect of anti-CD3 arm affinity on B cell depletion was analyzed.
  • FIGS. 10A and 10B show that the selected anti-CD3 antibodies had Thymidine incorporation reading on human and cynomolgus PBMC.
  • FIG. 11A-FIG. 11D show that all human anti-EpCam_h2B4 bispecific antibodies have cell killing activity on in vitro setting.
  • FIG. 12 shows that a single dose of human anti-BCMA/CD3 bispecific antibody resulted in tumor regression in a dose-dependent manner in an orthotopic MM1.S myeloma model.
  • FIG. 13 shows that two doses of human anti-BCMA/CD3 bispecific antibody resulted in increased tumor regression in an orthotopic Molp8 myeloma model.
  • FIG. 14 shows that anti-BCMA/CD3 bispecific antibody alone or in combination with standard of care for multiple myeloma (lenalidomide or bortezomib) is more efficacious than lenalidomide and bortezomib combined in orthotopic Molp8 tumor model.
  • FIG. 15A-FIG. 15C, respectively, show that carfilzomib, lenalidomide, and doxorubicin do not have a negative effect on the function of the anti-BCMA/CD3 bispecific antibody on OPM2 cells as compared to the anti-BCMA/CD3 bispecific antibody alone.
  • FIG. 16 shows synergistic effects on the function of anti-BCMA/CD3 bispecific antibody when combined with carfilzomib and lenalidomide in comparison to each molecule alone.
  • DETAILED DESCRIPTION
  • The invention disclosed herein provides antibodies and antibody conjugates (e.g., antibody-drug conjugates) that specifically bind to BCMA (e.g., human BCMA). The invention also provides polynucleotides encoding these antibodies and conjugates, compositions comprising these antibodies and conjugates, and methods of making these antibodies and conjugates. Further, the invention disclosed herein provides antibodies that specifically bind to CD3 (e.g., human CD3) as well as heterodimeric antibodies (e.g., bispecific antibodies) that specifically bind to CD3 and a tumor antigen (e.g., BCMA). The invention also provides polynucleotides encoding these antibodies, compositions comprising these antibodies, and methods of making and using these antibodies. The invention further provides methods for treating a condition associated with malignant BCMA expression in a subject, such as cancer or autoimmune disease, using the antibodies (e.g., BCMA, CD3, or bispecific antibody) or conjugates thereof (BCMA antibody-drug conjugates) as described herein.
  • General Techniques
  • The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al., 1989) Cold Spring Harbor Press; Oligonucleotide Synthesis (M. J. Gait, ed., 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J. E. Cellis, ed., 1998) Academic Press; Animal Cell Culture (R. I. Freshney, ed., 1987); Introduction to Cell and Tissue Culture (J. P. Mather and P. E. Roberts, 1998) Plenum Press; Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J. B. Griffiths, and D. G. Newell, eds., 1993-1998) J. Wiley and Sons; Methods in Enzymology (Academic Press, Inc.); Handbook of Experimental Immunology (D. M. Weir and C. C. Blackwell, eds.); Gene Transfer Vectors for Mammalian Cells (J. M. Miller and M. P. Calos, eds., 1987); Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Current Protocols in Immunology (J. E. Coligan et al., eds., 1991); Short Protocols in Molecular Biology (Wiley and Sons, 1999); Immunobiology (C. A. Janeway and P. Travers, 1997); Antibodies (P. Finch, 1997); Antibodies: a practical approach (D. Catty., ed., IRL Press, 1988-1989); Monoclonal antibodies: a practical approach (P. Shepherd and C. Dean, eds., Oxford University Press, 2000); Using antibodies: a laboratory manual (E. Harlow and D. Lane (Cold Spring Harbor Laboratory Press, 1999); The Antibodies (M. Zanetti and J. D. Capra, eds., Harwood Academic Publishers, 1995).
  • Definitions
  • An “antibody” is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. As used herein, the term encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab′, F(ab′)2, Fv), single chain (ScFv) and domain antibodies (including, for example, shark and camelid antibodies), and fusion proteins comprising an antibody, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site. An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant region of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. The heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • The term “antigen binding fragment” or “antigen binding portion” of an antibody, as used herein, refers to one or more fragments of an intact antibody that retain the ability to specifically bind to a given antigen (e.g., BCMA or CD3). Antigen binding functions of an antibody can be performed by fragments of an intact antibody. Examples of binding fragments encompassed within the term “antigen binding fragment” of an antibody include Fab; Fab′; F(ab′)2; an Fd fragment consisting of the VH and CH1 domains; an Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a single domain antibody (dAb) fragment (Ward et al., Nature 341:544-546, 1989), and an isolated complementarity determining region (CDR).
  • An antibody, an antibody conjugate, or a polypeptide that “preferentially binds” or “specifically binds” (used interchangeably herein) to a target (e.g., BCMA protein or CD3 protein) is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art. A molecule is said to exhibit “specific binding” or “preferential binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances. An antibody “specifically binds” or “preferentially binds” to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an antibody that specifically or preferentially binds to a BCMA epitope or CD3 epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other BCMA epitopes, non-BCMA epitopes, CD3 epitopes, or non-CD3 epitopes. It is also understood that by reading this definition, for example, an antibody (or moiety or epitope) that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target. As such, “specific binding” or “preferential binding” does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means preferential binding.
  • A “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. As known in the art, the variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions. The CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies. There are at least two techniques for determining CDRs: (1) an approach based on cross-species sequence variability (i.e., Kabat et al. Sequences of Proteins of Immunological Interest, (5th ed., 1991, National Institutes of Health, Bethesda MD)); and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al-lazikani et al., 1997, J. Molec. Biol. 273:927-948). As used herein, a CDR may refer to CDRs defined by either approach or by a combination of both approaches.
  • A “CDR” of a variable domain are amino acid residues within the variable region that are identified in accordance with the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art. Antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See, e.g., Kabat et al., 1992, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH, Washington D.C. The positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others. See, e.g., Chothia et al., Nature 342:877-883, 1989. Other approaches to CDR identification include the “AbM definition,” which is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys®), or the “contact definition” of CDRs based on observed antigen contacts, set forth in MacCallum et al., J. Mol. Biol., 262:732-745, 1996. In another approach, referred to herein as the “conformational definition” of CDRs, the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al., Journal of Biological Chemistry, 283:1156-1166, 2008. Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. As used herein, a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
  • As used herein, “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, Nature 256:495, 1975, or may be made by recombinant DNA methods such as described in U.S. Pat. No. 4,816,567. The monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et al., Nature 348:552-554, 1990, for example.
  • As used herein, “humanized” antibody refers to forms of non-human (e.g. murine) antibodies that are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab′, F(ab′)2 or other antigen binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin. Preferably, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementarity determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Preferred are antibodies having Fc regions modified as described in WO 99/58572. Other forms of humanized antibodies have one or more CDRs (CDR L1, CDR L2, CDR L3, CDR H1, CDR H2, or CDR H3) which are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody.
  • As used herein, “human antibody” means an antibody having an amino acid sequence corresponding to that of an antibody produced by a human and/or which has been made using any of the techniques for making human antibodies known to those skilled in the art or disclosed herein. This definition of a human antibody includes antibodies comprising at least one human heavy chain polypeptide or at least one human light chain polypeptide. One such example is an antibody comprising murine light chain and human heavy chain polypeptides. Human antibodies can be produced using various techniques known in the art. In one embodiment, the human antibody is selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., Nature Biotechnology, 14:309-314, 1996; Sheets et al., Proc. Natl. Acad. Sci. (USA) 95:6157-6162, 1998; Hoogenboom and Winter, J. Mol. Biol., 227:381, 1991; Marks et al., J. Mol. Biol., 222:581, 1991). Human antibodies can also be made by immunization of animals into which human immunoglobulin loci have been transgenically introduced in place of the endogenous loci, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. This approach is described in U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016. Alternatively, the human antibody may be prepared by immortalizing human B lymphocytes that produce an antibody directed against a target antigen (such B lymphocytes may be recovered from an individual or from single cell cloning of the cDNA, or may have been immunized in vitro). See, e.g., Cole et al. Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77, 1985; Boerner et al., J. Immunol., 147 (1):86-95, 1991; and U.S. Pat. No. 5,750,373.
  • The term “chimeric antibody” is intended to refer to antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody.
  • The terms “polypeptide”, “oligopeptide”, “peptide” and “protein” are used interchangeably herein to refer to chains of amino acids of any length, preferably, relatively short (e.g., 10-100 amino acids). The chain may be linear or branched, it may comprise modified amino acids, and/or may be interrupted by non-amino acids. The terms also encompass an amino acid chain that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It is understood that the polypeptides can occur as single chains or associated chains.
  • A “monovalent antibody” comprises one antigen binding site per molecule (e.g., IgG or Fab). In some instances, a monovalent antibody can have more than one antigen binding sites, but the binding sites are from different antigens.
  • A “monospecific antibody” comprises two identical antigen binding sites per molecule (e.g. IgG) such that the two binding sites bind identical epitope on the antigen. Thus, they compete with each other on binding to one antigen molecule. Most antibodies found in nature are monospecific. In some instances, a monospecific antibody can also be a monovalent antibody (e.g. Fab) A “bivalent antibody” comprises two antigen binding sites per molecule (e.g., IgG).
  • In some instances, the two binding sites have the same antigen specificities. However, bivalent antibodies may be bispecific.
  • A “bispecific” or “dual-specific” is a hybrid antibody having two different antigen binding sites. The two antigen binding sites of a bispecific antibody bind to two different epitopes, which may reside on the same or different protein targets.
  • A “bifunctional” is antibody is an antibody having identical antigen binding sites (i.e., identical amino acid sequences) in the two arms but each binding site can recognize two different antigens.
  • A “heteromultimer”, “heteromultimeric complex”, or “heteromultimeric polypeptide” is a molecule comprising at least a first polypeptide and a second polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue. The heteromultimer can comprise a “heterodimer” formed by the first and second polypeptide or can form higher order tertiary structures where polypeptides in addition to the first and second polypeptide are present.
  • A “heterodimer,” “heterodimeric protein,” “heterodimeric complex,” or “heteromultimeric polypeptide” is a molecule comprising a first polypeptide and a second polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue.
  • The “hinge region,” “hinge sequence”, and variations thereof, as used herein, includes the meaning known in the art, which is illustrated in, for example, Janeway et al., ImmunoBiology: the immune system in health and disease, (Elsevier Science Ltd., NY) (4th ed., 1999); Bloom et al., Protein Science (1997), 6:407-415; Humphreys et al., J. Immunol. Methods (1997), 209:193-202.
  • The “immunoglobulin-like hinge region,” “immunoglobulin-like hinge sequence,” and variations thereof, as used herein, refer to the hinge region and hinge sequence of an immunoglobulin-like or an antibody-like molecule (e.g., immunoadhesins). In some embodiments, the immunoglobulin-like hinge region can be from or derived from any IgG1, IgG2, IgG3, or IgG4 subtype, or from IgA, IgE, IgD or IgM, including chimeric forms thereof, e.g., a chimeric IgG1/2 hinge region.
  • The term “immune effector cell” or “effector cell as used herein refers to a cell within the natural repertoire of cells in the human immune system which can be activated to affect the viability of a target cell. The viability of a target cell can include cell survival, proliferation, and/or ability to interact with other cells.
  • Antibodies of the invention can be produced using techniques well known in the art, e.g., recombinant technologies, phage display technologies, synthetic technologies or combinations of such technologies or other technologies readily known in the art (see, for example, Jayasena, S. D., Clin. Chem., 45: 1628-50, 1999 and Fellouse, F. A., et al, J. Mol. Biol., 373(4):924-40, 2007).
  • As known in the art, “polynucleotide,” or “nucleic acid,” as used interchangeably herein, refer to chains of nucleotides of any length, and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a chain by DNA or RNA polymerase. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the chain. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. Other types of modifications include, for example, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide(s). Further, any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports. The 5′ and 3′ terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms. Other hydroxyls may also be derivatized to standard protecting groups. Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2′-O-methyl-, 2′-O-allyl, 2′-fluoro- or 2′-azido-ribose, carbocyclic sugar analogs, alpha- or beta-anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and abasic nucleoside analogs such as methyl riboside. One or more phosphodiester linkages may be replaced by alternative linking groups. These alternative linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S(“thioate”), P(S)S (“dithioate”), (O)NR2 (“amidate”), P(O)R, P(O)OR′, CO or CH2 (“formacetal”), in which each R or R′ is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (—O—) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • As known in the art a “constant region” of an antibody refers to the constant region of the antibody light chain or the constant region of the antibody heavy chain, either alone or in combination.
  • As used herein, “substantially pure” refers to material which is at least 50% pure (i.e., free from contaminants), more preferably, at least 90% pure, more preferably, at least 95% pure, yet more preferably, at least 98% pure, and most preferably, at least 99% pure.
  • A “host cell” includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts. Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. A host cell includes cells transfected in vivo with a polynucleotide(s) of this invention.
  • As known in the art, the term “Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain. The “Fc region” may be a native sequence Fc region or a variant Fc region. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The numbering of the residues in the Fc region is that of the EU index as in Kabat. Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991. The Fc region of an immunoglobulin generally comprises two constant regions, CH2 and CH3.
  • As used in the art, “Fc receptor” and “FcR” describe a receptor that binds to the Fc region of an antibody. The preferred FcR is a native sequence human FcR. Moreover, a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcγRI, FcγRII, and FcγRIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcγRII receptors include FcγRIIA (an “activating receptor”) and FcγRIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. FcRs are reviewed in Ravetch and Kinet, Ann. Rev. Immunol., 9:457-92, 1991; Capel et al., Immunomethods, 4:25-34, 1994; and de Haas et al., J. Lab. Clin. Med., 126:330-41, 1995. “FcR” also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol., 117:587, 1976; and Kim et al., J. Immunol., 24:249, 1994).
  • The term “compete”, as used herein with regard to an antibody, means that a first antibody, or an antigen binding fragment (or portion) thereof, binds to an epitope in a manner sufficiently similar to the binding of a second antibody, or an antigen binding portion thereof, such that the result of binding of the first antibody with its cognate epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody. The alternative, where the binding of the second antibody to its epitope is also detectably decreased in the presence of the first antibody, can, but need not be the case. That is, a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope. However, where each antibody detectably inhibits the binding of the other antibody with its cognate epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to “cross-compete” with each other for binding of their respective epitope(s). Both competing and cross-competing antibodies are encompassed by the present invention. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate, based upon the teachings provided herein, that such competing and/or cross-competing antibodies are encompassed and can be useful for the methods disclosed herein.
  • A “functional Fc region” possesses at least one effector function of a native sequence Fc region. Exemplary “effector functions” include Clq binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity; phagocytosis; down-regulation of cell surface receptors (e.g. B cell receptor), etc. Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
  • A “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature. A “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region. In some embodiments, the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably, from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide. The variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably, at least about 90% sequence identity therewith, more preferably, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% sequence identity therewith.
  • The term “effector function” refers to the biological activities attributable to the Fc region of an antibody. Examples of antibody effector functions include, but are not limited to, antibody-dependent cell-mediated cytotoxicity (ADCC), Fc receptor binding, complement dependent cytotoxicity (CDC), phagocytosis, Clq binding, and down regulation of cell surface receptors (e.g., B cell receptor; BCR). See, e.g., U.S. Pat. No. 6,737,056. Such effector functions generally require the Fc region to be combined with a binding domain (e.g., an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions. An exemplary measurement of effector function is through Fcγ3 and/or Clq binding.
  • As used herein “antibody-dependent cell-mediated cytotoxicity” or “ADCC” refers to a cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g. natural killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. ADCC activity of a molecule of interest can be assessed using an in vitro ADCC assay, such as that described in U.S. Pat. No. 5,500,362 or 5,821,337. Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and NK cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes et al., 1998, PNAS (USA), 95:652-656.
  • “Complement dependent cytotoxicity” or “CDC” refers to the lysing of a target in the presence of complement. The complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule (e.g. an antibody) complexed with a cognate antigen. To assess complement activation, a CDC assay, e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods, 202: 163 (1996), may be performed.
  • As used herein, “treatment” is an approach for obtaining beneficial or desired clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cells, inhibiting metastasis of neoplastic cells, remission of a BCMA associated disease (e.g., cancer or autoimmune disease), decreasing symptoms resulting from a BCMA associated disease (e.g., cancer or autoimmune disease), increasing the quality of life of those suffering from a BCMA associated disease (e.g., cancer or autoimmune disease), decreasing the dose of other medications required to treat a BCMA associated disease (e.g., cancer or autoimmune disease), delaying the progression of a BCMA associated disease (e.g., cancer or autoimmune disease), curing a BCMA associated disease (e.g., cancer or autoimmune disease), and/or prolong survival of patients having a BCMA associated disease (e.g., cancer or autoimmune disease). “Ameliorating” means a lessening or improvement of one or more symptoms as compared to not administering a BCMA antibody or a BCMA antibody conjugate. “Ameliorating” also includes shortening or reduction in duration of a symptom.
  • As used herein, an “effective dosage” or “effective amount” of drug, compound, or pharmaceutical composition is an amount sufficient to effect any one or more beneficial or desired results. For prophylactic use, beneficial or desired results include eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • For therapeutic use, beneficial or desired results include clinical results such as reducing incidence or amelioration of one or more symptoms of various BCMA associated diseases or conditions (such as multiple myeloma), decreasing the dose of other medications required to treat the disease, enhancing the effect of another medication, and/or delaying the progression of the BCMA associated disease of patients. An effective dosage can be administered in one or more administrations. For purposes of this invention, an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. As is understood in the clinical context, an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • An “individual” or a “subject” is a mammal, more preferably, a human. Mammals also include, but are not limited to, farm animals, sport animals, pets, primates, horses, dogs, cats, mice and rats.
  • As used herein, “vector” means a construct, which is capable of delivering, and, preferably, expressing, one or more gene(s) or sequence(s) of interest in a host cell.
  • Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells.
  • As used herein, “expression control sequence” means a nucleic acid sequence that directs transcription of a nucleic acid. An expression control sequence can be a promoter, such as a constitutive or an inducible promoter, or an enhancer. The expression control sequence is operably linked to the nucleic acid sequence to be transcribed.
  • As used herein, “pharmaceutically acceptable carrier” or “pharmaceutical acceptable excipient” includes any material which, when combined with an active ingredient, allows the ingredient to retain biological activity and is non-reactive with the subject's immune system. Examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents. Preferred diluents for aerosol or parenteral administration are phosphate buffered saline (PBS) or normal (0.9%) saline. Compositions comprising such carriers are formulated by well known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, P A, 1990; and Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005).
  • The term “acyl donor glutamine-containing tag” or “glutamine tag” as used herein refers to a polypeptide or a protein containing one or more Gln residue(s) that acts as a transglutaminase amine acceptor. See, e.g., WO2012059882 and WO2015015448.
  • The term “kon” or “ka”, as used herein, refers to the rate constant for association of an antibody to an antigen. Specifically, the rate constants (kon/ka and koff/kd) and equilibrium dissociation constants are measured using whole antibody (i.e. bivalent) and monomeric BCMA proteins.
  • The term “kon” or “kd”, as used herein, refers to the rate constant for dissociation of an antibody from the antibody/antigen complex.
  • The term “KD”, as used herein, refers to the equilibrium dissociation constant of an antibody-antigen interaction.
  • Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X.” Numeric ranges are inclusive of the numbers defining the range.
  • It is understood that wherever embodiments are described herein with the language “comprising,” otherwise analogous embodiments described in terms of “consisting of” and/or “consisting essentially of” are also provided.
  • Where aspects or embodiments of the invention are described in terms of a Markush group or other grouping of alternatives, the present invention encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group, but 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 claimed invention.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In case of conflict, the present specification, including definitions, will control. Throughout this specification and claims, the word “comprise,” or variations such as “comprises” or “comprising” will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers. Unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.
  • Exemplary methods and materials are described herein, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention. The materials, methods, and examples are illustrative only and not intended to be limiting.
  • BCMA Antibodies and Methods of Making Thereof
  • The present invention provides an antibody that binds to BCMA (e.g., human BCMA (e.g., SEQ ID NO: 353 or accession number: Q02223-2) and characterized by any one or more of the following characteristics: (a) treat, prevent, ameliorate one or more symptoms of a condition associated with malignant cells expressing BCMA in a subject (e.g., B-cell related cancer such as multiple myeloma); (b) inhibit tumor growth or progression in a subject (who has a malignant tumor expressing BCMA); (c) inhibit metastasis of cancer (malignant) cells expressing BCMA in a subject (who has one or more malignant cells expressing BCMA); (f) induce regression (e.g., long-term regression) of a tumor expressing BCMA; (d) exert cytotoxic activity in malignant cells expressing BCMA; and (e) block BCMA interaction with other yet to be identified factors.
  • In one aspect, provided is an isolated antibody, or an antigen binding fragment thereof, which specifically binds to B-Cell Maturation Antigen (BCMA), wherein the antibody comprises (a) a heavy chain variable (VH) region comprising (i) a VH complementary determining region one (CDR1) comprising the sequence SYX1MX2, wherein X1 is A or P; and X2 is T, N, or S (SEQ ID NO: 301), GFTFX1SY, wherein X1 is G or S (SEQ ID NO: 302), or GFTFX1SYX2MX3, wherein X1 is G or S, X2 is A or P; and X3 is T, N, or S (SEQ ID NO: 303); (ii) a VH CDR2 comprising the sequence AX1X2X3X4GX5X6X7X8YADX9X10KG, wherein X1 is I, V, T, H, L, A, or C; X2 is S, D, G, T, I, L, F, M, or V; X3 is G, Y, L, H, D, A, S, or M; X4 is S, Q, T, A, F, or W; X5 is G or T; X6 is N, S, P, Y, W, or F; X7 is S, T, I, L, T, A, R, V, K, G, or C; X8 is F, Y, P, W, H, or G; X9 is V, R, or L; and X10 is G or T (SEQ ID NO: 305), or X1X2X3X4X5X6, wherein X1 is S, V, I, D, G, T, L, F, or M; X2 is G, Y, L, H, D, A, S, or M; X3 is S, G, F, or W; X4 is G or S; X5 is G or T; and X6 is N, S, P, Y, or W (SEQ ID NO: 306); and iii) a VH CDR3 comprising the sequence VSPIX1X2X3X4, wherein X1 is A or Y; X2 is A or S; and X3 is G, Q, L, P, or E (SEQ ID NO: 307), or YWPMX1X2, wherein X1 is D, S, T, or A; and X2 is I, S, L, P, or D (SEQ ID NO: 308); and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence X1X2X3X4X5X6X7X8X9X10X11X12, wherein X1 is R, G, W, A, or C; X2 is A, P, G, L, C, or S; X3 is S, G, or R; X4 is Q, C, E, V, or I; X5 is S, P, G, A, R, or D; X6 is V, G, I, or L; X7 is S, E, D, P, or G; X8 is S, P, F, A, M, E, V, N, D, or Y; X9 is I, T, V, E, S, A, M, Q, Y, H, R, or F; X10 is Y or F; X11 is L, W, or P; and X12 is A, S, or G (SEQ ID NO: 309); (ii) a VL CDR2 comprising the sequence X1ASX2RAX3, wherein X1 is G or D; X2 is S or I; and X3 is T or P (SEQ ID NO: 310); and (iii) a VL CDR3 comprising the sequence QQYX1X2X3PX4T, wherein X1 is G, Q, E, L, F, A, S, M, K, R, or Y; X2 is S, R, T, G, V, F, Y, D, A, H, V, E, K, or C; X3 is W, F, or S; and X4 is L or I (SEQ ID NO: 311), or QQYX1X2X3PX4, wherein X1 is G, Q, E, L, F, A, S, M, R, K, or Y; X2 is S, R, T, G, R, V, D, A, H, E, K, C, F, or Y; X3 is W, S, or F; and X4 is L or I (SEQ ID NO: 312).
  • In another aspect, provided is an isolated antibody, or an antigen binding fragment thereof, which specifically binds to BCMA, wherein the antibody comprises: a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 112, 125, 127, 313, 314, 363, or 365; and/or a VL region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38, 40, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 317, 80, 81, 82, 84, 85, 86, 88, 89, 90, 91, 93, 94, 96, 98, 100, 102, 103, 105, 107, 108, 109, 111, 113, 115, 116, 117, 119, 121, 123, 124, 126, 128, 315, 316, or 364.
  • In some embodiments, provided is an antibody having any one of partial light chain sequence as listed in Table 1 and/or any one of partial heavy chain sequence as listed in Table 1.
  • TABLE 1
    mAb Light Chain Heavy Chain
    P6E01/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    P6E01 ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YGSPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 1) VSS (SEQ ID NO: 2)
    P6E01/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H3.AQ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YGSPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 1) VSS (SEQ ID NO: 3)
    L1.LG EIVLTQSPGTLSLSPGERATLSC EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K RASQSLGSFYLA WYQQKPGQA ASGFTFG SYAMTWVRQAPGKGLE
    W/P6E PRLLIY GASSRAT GIPDRFSGSG WVSAISGSGGNTFYADSVKGRFTI
    01 SGTDFTLTISRLEPEDFAVYYC KH SRDNSKNTLYLQMNSLRAEDTAV
    YGWPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 4) VSS (SEQ ID NO: 2)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/P6E RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    01 GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 5) VSS (SEQ ID NO: 2)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/P6E RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    01 GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 6) VSS (SEQ ID NO: 2)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    W/H3. RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    AL GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR ARVSPIAALMDY WGQGTL
    (SEQ ID NO: 4) VTVSS (SEQ ID NO: 7)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    W/H3. RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    AP GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR VSPIAAPMDY WGQGTLVT
    (SEQ ID NO: 4) VSS (SEQ ID NO: 8)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    W/H3. RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    AQ GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 4) VSS (SEQ ID NO: 3)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.P ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    P GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAPMDY WGQGTLVT
    (SEQ ID NO: 9) VSS (SEQ ID NO: 8)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.P ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 9) VSS (SEQ ID NO: 3)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    L GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIAALMDY WGQGTLVT
    (SEQ ID NO: 10) VSS (SEQ ID NO: 7)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    P GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIAAPMDY WGQGTLVT
    (SEQ ID NO: 10) VSS (SEQ ID NO: 8)
    L1.LG EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSLGSFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 10) VSS (SEQ ID NO: 3)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    W/H3. RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    AL GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR VSPIAALMDY WGQGTLVT
    (SEQ ID NO: 11) VSS (SEQ ID NO: 7)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    W/H3. RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    AP GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR VSPIAAPMDY WGQGTLVT
    (SEQ ID NO: 11) VSS (SEQ ID NO: 8)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.K ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    W/H3. RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    AQ GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 11) VSS (SEQ ID NO: 3)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.P ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 12) VSS (SEQ ID NO: 3)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    L GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIAALMDY WGQGTLVT
    (SEQ ID NO: 13) VSS (SEQ ID NO: 7)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    P GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIAAPMDY WGQGTLVT
    (SEQ ID NO: 13) VSS (SEQ ID NO: 8)
    L1.GD EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    F/L3.N ASQSVGDFYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Y/H3.A RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 14) VSS (SEQ ID NO: 3)
    L3.KW/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    P6E01 ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KHY SRDNSKNTLYLQMNSLRAEDTAV
    GWPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 15) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    P6E01 ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 2)
    L3.NY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    P6E01 ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YNYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 17) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    P6E01 RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    P6E01 RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    P6E01 LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PH/ ASQSVSPHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    P6E01 RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 21) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    P6E01 RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    P6E01 RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H2.QR ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQRKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 24)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H2.DY ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAIDYSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 25)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H2.YQ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISYQGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 26)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H2.LT ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISLTGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 27)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H2.HA ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISHAGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 28)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H2.QL ASQSVSSSYLA WYQQKPGQAPRLLI ASGFTFG SYAMTWVRQAPGKGLE
    Y GASSRAT GIPDRFSGSGSGTDFT WVSAISGSGGNTFYADQLKGRFTI
    LTISRLEPEDFAVYYC QHYPYPPSFT SRDNSKNTLYLQMNSLRAEDTAV
    FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 29)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H3.YA ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIYAGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 30)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H3.AE ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAEMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 31)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H3.AQ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 3)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    H3.TA ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    Q RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 32)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    P6E01 ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 16) VSS (SEQ ID NO: 2)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QR RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQRKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 24)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.DY RLLIY GASSRAT GIPDRFSGSGS WVSAIDYSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 25)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.YQ RLLIY GASSRAT GIPDRFSGSGS WVSAISYQGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 26)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.LT RLLIY GASSRAT GIPDRFSGSGS WVSAISLTGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 27)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.HA RLLIY GASSRAT GIPDRFSGSGS WVSAISHAGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 28)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QL RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQLKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 29)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.YA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIYAGMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 30)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AE RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAEMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 31)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AQ RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 3)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PS/ ASQSVSSSYPS WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.TA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 18) VSS (SEQ ID NO: 32)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QR RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQRKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 24)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.DY RLLIY GASSRAT GIPDRFSGSGS WVSAIDYSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 25)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.YQ RLLIY GASSRAT GIPDRFSGSGS WVSAISYQGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 26)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.LT RLLIY GASSRAT GIPDRFSGSGS WVSAISLTGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 27)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.HA RLLIY GASSRAT GIPDRFSGSGS WVSAISHAGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 28)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QL RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQLKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 29)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.YA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIYAGMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 30)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AE RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAEMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 31)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AQ RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 3)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.AH/ ASQSVSAHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.TA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 19) VSS (SEQ ID NO: 32)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H2.QR LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADQRKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 24)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H2.DY LLIY GASSRAT GIPDRFSGSGSG WVSAIDYSGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 25)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H2.YQ LLIY GASSRAT GIPDRFSGSGSG WVSAISYQGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 26)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H2.LT LLIY GASSRAT GIPDRFSGSGSG WVSAISLTGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 27)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H2.HA LLIY GASSRAT GIPDRFSGSGSG WVSAISHAGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 28)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H2.QL LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADQLKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 29)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H3.YA LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIYAGMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 30)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H3.AE LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIAAEMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 31)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H3.AQ LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 3)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.FF/ ASQSVSSFFLA WYQQKPGQAPR ASGFTFG SYAMTWVRQAPGKGLE
    H3.TA LLIY GASSRAT GIPDRFSGSGSG WVSAISGSGGNTFYADSVKGRFTI
    Q TDFTLTISRLEPEDFAVYYC QHYP SRDNSKNTLYLQMNSLRAEDTAV
    YPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 20) VSS (SEQ ID NO: 32)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PH/ ASQSVSPHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QR RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQRKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 21) VSS (SEQ ID NO: 24)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PH/ ASQSVSPHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.HA RLLIY GASSRAT GIPDRFSGSGS WVSAISHAGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 21) VSS (SEQ ID NO: 28)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PH/ ASQSVSPHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AE RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAEMDY WGQGTLVT
    (SEQ ID NO: 21) VSS (SEQ ID NO: 31)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PH/ ASQSVSPHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AQ RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 21) VSS (SEQ ID NO: 3)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L1.PH/ ASQSVSPHYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.TA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YPYPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 21) VSS (SEQ ID NO: 32)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QR RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQRKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 24)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.DY RLLIY GASSRAT GIPDRFSGSGS WVSAIDYSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 25)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.YQ RLLIY GASSRAT GIPDRFSGSGS WVSAISYQGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 26)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.LT RLLIY GASSRAT GIPDRFSGSGS WVSAISLTGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 27)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.HA RLLIY GASSRAT GIPDRFSGSGS WVSAISHAGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 28)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QL RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQLKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 29)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.YA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIYAGMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 30)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KY/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.TA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC KYY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 22) VSS (SEQ ID NO: 32)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.DY RLLIY GASSRAT GIPDRFSGSGS WVSAIDYSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 25)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.YQ RLLIY GASSRAT GIPDRFSGSGS WVSAISYQGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 26)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.LT RLLIY GASSRAT GIPDRFSGSGS WVSAISLTGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 27)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H2.QL RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADQLKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIASGMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 29)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.YA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIYAGMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 30)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AE RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIAAEMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 31)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.AQ RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCAR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 3)
    L3.PY/ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    L3.KF/ ASQSVSSSYLA WYQQKPGQAP ASGFTFG SYAMTWVRQAPGKGLE
    H3.TA RLLIY GASSRAT GIPDRFSGSGS WVSAISGSGGNTFYADSVKGRFTI
    Q GTDFTLTISRLEPEDFAVYYC KFY SRDNSKNTLYLQMNSLRAEDTAV
    PYPPSFT FGQGTKVEIK  YYCTR VSPIAAQMDY WGQGTLVT
    (SEQ ID NO: 23) VSS (SEQ ID NO: 32)
    P5A2_ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    VHVL ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    RLLMY DASIRAT GIPDRFSGSGS WVSAI SDSGGS TYYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YGSWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 34) (SEQ ID NO: 33)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSVIYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    nM_C0 LLMY DASIRAT GIPDRFSGSGSG WVSAI SDSGGS AWYADSVKGRFT
    6 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    QRWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 36) (SEQ ID NO: 35)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    nM_C0 RLLMY DASIRAT GIPDRFSGSGS WVSAI SDSGGS MWYADSVKGRF
    9 GTDFTLTISRLEPEDFAVYYC QQ TISRDNSKNTLYLQMNSLRAEDTA
    YQSWPLT FGQGTKVEIK  VYYCARYWPMSLWGQGTLVTVS
    (SEQ ID NO: 38) S (SEQ ID NO: 37)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSDIYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    M_C16 LLMY DASIRAT GIPDRFSGSGSG WVSAI SdFGGS TYYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    QTWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 40) (SEQ ID NO: 39)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSNLYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C03 RLLMY DASIRAT GIPDRFSGSGS WVSAI SDSGGS TYYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YQGWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 41) (SEQ ID NO: 33)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSAYYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C01 RLLMY DASIRAT GIPDRFSGSGS WVSAI TASGGS TYYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YERWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 43) (SEQ ID NO: 42)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSSLYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    M_C26 LLMY DASIRAT GIPDRFSGSGSG WVSAI SDSGGS TYYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    QVWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 45) (SEQ ID NO: 44)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C25 RLLMY DASIRAT GIPDRFSGSGS WVSAI SdSGGS RWYADSVKGRFT
    GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YLDWPLT FGQGTKVEIK  YYCAR YWPMTP WGQGTLVTVSS
    (SEQ ID NO: 47) (SEQ ID NO: 46)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C22 RLLMY DASIRAT GIPDRFSGSGS WVSAV LdSGGS TYYADSVKGRFT
    GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YQVWPLT FGQGTKVEIK  YYCAR YWPMTP WGQGTLVTVSS
    (SEQ ID NO: 49) (SEQ ID NO: 48)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSVIYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    M_C19 LLMY DASIRAT GIPDRFSGSGSG WVSAI SdSGGS RWYADSVKGRFT
    TDFTLTISRLEPEDFAVYYC QQYL ISRDNSKNTLYLQMNSLRAEDTAV
    AWPLT FGQGTKVEIK  YYCAR YWPMSD WGQGTLVTVSS
    (SEQ ID NO: 51) (SEQ ID NO: 50)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    nM_CO RLLMY DASIRAT GIPDRFSGSGS WVSAI SdSGGS KWYADSVKGRFT
    3 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YFTWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 53) (SEQ ID NO: 52)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSPyYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    M_C07 LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    TDFTLTISRLEPEDFAVYYC QQYE ISRDNSKNTLYLQMNSLRAEDTAV
    RWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 55) (SEQ ID NO: 54)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSVEYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C23 RLLMY DASIRAT GIPDRFSGSGS WVSAI SdSGGS GWYADSVKGRFT
    GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YARWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 57) (SEQ ID NO: 56)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSEIYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    nM_C1 LLMY DASIRAT GIPDRFSGSGSG WVSAV LdSGGS TYYADSVKGRFT
    8 TDFTLTISRLEPEDFAVYYC QQYF ISRDNSKNTLYLQMNSLRAEDTAV
    GWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 59) (SEQ ID NO: 58)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVEMSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C10 RLLMY DASIRAT GIPDRFSGSGS WVSAI SdSGGS CWYADSVKGRFT
    GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YAHWPLT FGQGTKVEIK  YYCAR YWPMTP WGQGTLVTVSS
    (SEQ ID NO: 61) (SEQ ID NO: 60)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    M_C05 RLLMY DASIRAT GIPDRFSGSGS WVSAI FaSGGS TYYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YQRWPLT FGQGTKVEIK  YYCAR YWPMTP WGQGTLVTVSS
    (SEQ ID NO: 63) (SEQ ID NO: 62)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSAQYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    nM_C1 RLLMY DASIRAT GIPDRFSGSGS WVSAI SgWGGS LPYADSVKGRFT
    0 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YQRWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 65) (SEQ ID NO: 64)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSAIYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    M_C04 LLMY DASIRAT GIPDRFSGSGSG WVSAI MsSGGP LYYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    QVWPLT FGQGTKVEIK  YYCAR YWPMAL WGQGTLVTVSS
    (SEQ ID NO: 67) (SEQ ID NO: 66)
    A02_R EIVLTQSPGTLSLSPGERATLSC G EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 PSQSVSSSYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    nM_C2 LLMY DASIRAT GIPDRFSGSGSG WVSAI LmSGGS TYYADSVKGRFTI
    6 TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    QSWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 69) (SEQ ID NO: 68)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSSSYWA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    nM_C1 RLLMY DASIRAT GIPDRFSGSGS WVSAI SdSGGY RYYADSVKGRFTI
    3 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YESWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 71) (SEQ ID NO: 70)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 GGQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    nM_C0 RLLMY DASIRAT GIPDRFSGSGS WVSAI LsSGGS TYYADSVKGRFTI
    1 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YQSWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 73) (SEQ ID NO: 72)
    A02_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSFIYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    M_C08 LLMY DASIRAT GIPDRFSGSGSG WVSAI LdSGGS TYYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    GSWPLT FGQGTKVEIK  YYCAR YWPMSP WGQGTLVTVSS
    (SEQ ID NO: 75) (SEQ ID NO: 74)
    P5C1_ EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    VHVL ASQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    LLIY DASSRAP GIPDRFSGSGSG WVSAI GGSGGS TYYADSVKGRFT
    TDFTLTISRLEPEDFAVYYC QQYS ISRDNSKNTLYLQMNSLRAEDTAV
    TSPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 77) (SEQ ID NO: 76)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSPEYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    M_C24 RLLIY DASSRAP GIPDRFSGSGS WVSAI GGSGGS LPYADSVKGRFT
    GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YSVWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 79) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSAIYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    M_C26 LLIY DASSRAP GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    TDFTLTISRLEPEDFAVYYC QQYS ISRDNSKNTLYLQMNSLRAEDTAV
    AWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 317) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSSvYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    M_C10 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    TWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 79) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    nM_C2 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    7 TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    RWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 81) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_6n ASQSVSPIYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    M_C20 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    AFPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 82) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC EVQLLESGGGLVQPGGSLRLSCA
    d4_6n WLSQSVSSTYLA WYQQKPGQA ASGFTFS SYPMSWVRQAPGKGLE
    M_C12 PRLLIY DASSRAP GIPDRFSGSG WVSAI GgSGGW SYYADSVKGRFT
    SGTDFTLTISRLEPEDFAVYYC Q ISRDNSKNTLYLQMNSLRAEDTAV
    QYSEWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 84) (SEQ ID NO: 83)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    nM_C1 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    6 TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    SWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 85) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASQSVSSIFLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    nM_C0 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    9 TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    AWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 86) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC A EVQLLESGGGLVQPGGSLRLSCA
    d4_6n CSQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    M_C09 LLIY DASSRAP GIPDRFSGSGSG WVSAT VgSGGS IGYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    AWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 88) (SEQ ID NO: 87)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASCDVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    nM_C0 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    3 TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    MRSPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 89) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    d4_0.6 ASEAVPSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    nM_C0 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGTISR
    6 TDFTLTISRLEPEDFAVYYC QQYS DNSKNTLYLQMNSLRAEDTAVYY
    AFPLT FGQGTKVEIK  CAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 90) (SEQ ID NO: 78)
    C01_R EIVLTQSPGTLSLSPGERATLSC C EVQLLESGGGLVQPGGSLRLSCA
    d4_6n SSQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    M_C04 LLIY DASSRAP GIPDRFSGSGSG WVSAI GgSGGS LPYADSVKGRFTI
    TDFTLTISRLEPEDFAVYYC QQYS SRDNSKNTLYLQMNSLRAEDTAV
    AFPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 91) (SEQ ID NO: 78)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASVRVSSTYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    _0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAI SdSGGS RWYADSVKGRFT
    M_C22 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    MKWPLT FGQGTKVEIK  YYCTR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 93) (SEQ ID NO: 92)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSAAYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAI GgSGGS LPYADSVKGRFTI
    C21 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YMCWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 94) (SEQ ID NO: 78)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSSYWG WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAI GgSGGS IHYADSVKGRFTI
    C10 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YQCWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 96) (SEQ ID NO: 95)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAH IgSGGS TYYADSVKGRFTI
    M_C04 TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    QSWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 98) (SEQ ID NO: 97)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSpYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ LLMY DASIRAT GIPDRFSGSGSG WVSAI GgSGGS TYYADSVKGRFTI
    C25 TDFTLTISRLEPEDFAVYYC QQY SRDNSKNTLYLQMNSLRAEDTAV
    QSWPLT FGQGTKVEIK  YYCAR YWPMDP WGQGTLVTVSS
    (SEQ ID NO: 100) (SEQ ID NO: 99)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLMY DASIRAT GIPDRFSGSGS WVSAI GgSGGS LPYADSVKGRFTI
    M_C21 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YQSWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 38) (SEQ ID NO: 78)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSPIYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LGYADSVKGRFT
    C11 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    KAWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 102) (SEQ ID NO: 101)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSYLYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    M_C20 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    MEWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 103) (SEQ ID NO: 78)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSAQYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAI FASGGS TYYADSVKGRFTI
    C09 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YQAWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 105) (SEQ ID NO: 104)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAI GGSGTW TYYADSVKGRFT
    C08 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YQKWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 107) (SEQ ID NO: 106)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSAVYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLMY DASIRAT GIPDRFSGSGS WVSAI GGSGGS LPYADSVKGRFT
    M_C19 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YRAWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 108) (SEQ ID NO: 78)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASIAVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    M_C02 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    MVWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 109) (SEQ ID NO: 78)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 PRQSVSSSYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLMY DASIRAT GIPDRFSGSGS WVSAL FGSGGS TYYADSVKGRFT
    M_C23 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YQDWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 111) (SEQ ID NO: 110)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLMY DASIRAT GIPDRFSGSGS WVSAI GGSGGS LPYADSVKGRFT
    M_C29 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YQSWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 38) (SEQ ID NO: 112)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    M_C09 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    QEWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 113) (SEQ ID NO: 112)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSASYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAA LGSGGS TYYADSVKGRF
    C12 GTDFTLTISRLEPEDFAVYYC QQ TISRDNSKNTLYLQMNSLRAEDTA
    YMSWPLT FGQGTKVEIK  VYYCAR YWPMDS WGQGTLVTVS
    (SEQ ID NO: 115) S (SEQ ID NO: 114)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSYMYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLIY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS TYYADSVKGRFT
    M_C30 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    KSWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 116) (SEQ ID NO: 76)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSALYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLMYDASIRATGIPDRFSGSGS WVSAI GGSGGS LPYADSVKGRFT
    M_C14 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YYGWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 117) (SEQ ID NO: 112)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQPISSSYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _6nM_ LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    C07 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    QGWPLT FGQGTKVEIK  YYCAR YWPMAD WGQGTLVTVSS
    (SEQ ID NO: 119) (SEQ ID NO: 118)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAI SDSGGF VYYADSVKGRFTI
    C02 GTDFTLTISRLEPEDFAVYYC QQ SRDNSKNTLYLQMNSLRAEDTAV
    YEFWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 121) (SEQ ID NO: 120)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSTYLA WYQQKPGQAPR ASGFTFS SYAMNWVRQAPGKGLE
    _0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS TYYADSVKGRFT
    M_C05 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    MSWPLT FGQGTKVEIK  YYCAR YWPMSL WGQGTLVTVSS
    (SEQ ID NO: 123) (SEQ ID NO: 122)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQGISSTYLA WYQQKPGQAPR ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n LLMY DASIRAT GIPDRFSGSGSG WVSAI GGSGGS LPYADSVKGRFT
    M_C17 TDFTLTISRLEPEDFAVYYC QQY ISRDNSKNTLYLQMNSLRAEDTAV
    AYWPLT FGQGTKVEIK  YYCAR YWPMDI WGQGTLVTVSS
    (SEQ ID NO: 124) (SEQ ID NO: 112)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMNWVRQAPGKGLE
    _6nM_ RLLMY DASIRAT GIPDRFSGSGS WVSAC LDSGGS TYYADSVKGRFT
    C22 GTDFTLTISRLEPEDFAVYYC QQ ISRDNSKNTLYLQMNSLRAEDTAV
    YQGWPLT FGQGTKVEIK  YYCAR YWPMDS WGQGTLVTVSS
    (SEQ ID NO: 126) (SEQ ID NO: 125)
    COMB EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    O_Rd4 ASQSVSVRYLA WYQQKPGQAP ASGFTFS SYPMSWVRQAPGKGLE
    _0.6n RLLMY DASIRAT GIPDRFSGSGS WVSAA LGSGGS TYYADSVKGRF
    M_C11 GTDFTLTISRLEPEDFAVYYC QQ TISRDNSKNTLYLQMNSLRAEDTA
    YGSWPIT FGQGTKVEIK  VYYCAR YWPMSL WGQGTLVTVS
    (SEQ ID NO: 128) S (SEQ ID NO: 127)
    Con- EIVLTQSPGTLSLSPGERATLSC EVQLLESGGGLVQPGGSLRLSCA
    sensus X1X2X3X4X5X6X7X8X9X10X11X12WY ASGFTFX1SYX2MX3WVRQAPGKG
    QQKPGQAPRLLMYX13ASX14RAX15 LEWVSAX4X5X6X7GX8X9X10X11YAD
    GIPDRFSGSGSGTDFTLTISRLE X12X13KGRFTISRDNSKNTLYLQMN
    PEDFAVYYCX16X17YX18X19PPSF SLRAEDTAVYYCARVSPIX14X15X16
    TFGQGTKVEIK, wherein X1 is R, MDYWGQGTLVTVSS, wherein X1
    G, W, A, or C; X2 is A, P, G,  is G or S, X2 is A or P; X3
    L, C, or S; X3 is S, G, or R; is T, N, or S; X4 is I, V,
    X4 is Q, C, E, V, or I; X5 is T, H, L, A, or C; X5 is S,
    S, P, G, A, R, or D; X6 is V, D, G, T, I, L, F, M, or V; 
    G, I, or L; X7 is S, E, D, P, X6 is G, Y, L, H, D, A, S,
    or G; X8 is S, P, F, A, M, E,  or M; X7 is S, Q, T, A, F,
    V, N, D, or Y; X9 is I, T, V, or W; X8 is G or T; X9 is N, 
    E, S, A, M, Q, Y, H, R, or F; S, P, Y, W, or F; X10 is S,
    X10 is Y or F; X11 is L, W, T, I, L, T, A, R, V, K, G,
    or P; X12 is A, S, or G, X13 or C; X11 is F, Y, P, W, H,
    is G or D; X14 is S or I; X15 or G; X12 is V, R, or L; X13
    is T or P; X16 is Q or K; X17 is G or T; X14 is A or Y; X15
    is H or Y; X18 is G, N, or P; is A or S; and X16 is G, Q, 
    and X19 is S, W, or Y L, P, or E 
    (SEQ ID NO: 315); or (SEQ ID NO: 313); or
    EIVLTQSPGTLSLSPGERATLSC EVQLLESGGGLVQPGGSLRLSCA
    X1X2X3X4X5X6X7X8X9X10X11X12WY ASGFTFX1SYX2MX3WVRQAPGKG
    QQKPGQAPRLLMYX13ASX14RAX15 LEWVSAX4X5X6X7GX8X9X10X11YAD
    GIPDRFSGSGSGTDFTLTISRLE X12X13KGRFTISRDNSKNTLYLQMN
    PEDFAVYYCQQYX16X17X18PX19F SLRAEDTAVYYCARYWPMX14X15
    GQGTKVEIK, wherein X1 is R, WGQGTLVTVSS, wherein X1 is 
    G, W, A, or C; X2 is A, P, G, G or S, X2 is A or P; X3 is
    L, C, or S; X3 is S, G, or R; T, N, or S; X4 is I, V, T,  
    X4 is Q, C, E, V, or I; X5 is H, L, A, or C; X5 is S, D,
    S, L, P, G, A, R, or D; X6 is G, T, I, L, F, M, or V; X6
    V, G, or I; X7 is S, E, D, or  is G, Y, L, H, D, A, S, or
    P; X8 is S, P, F, A, M, E, V, M; X7 is S, Q, T, A, F, or
    N, D, or Y; X9 is I, T, V, E,  W; X8 is G or T; X9 is N,  
    S, A, M, Q, Y, H, or R; X10 S, P, Y, W, or F; X10 is
    is Y or F; X11 is L, W, or P; S, T, I, L, T, A, R, V, K,
    X12 is A, S, or G, X13 is G  G, or C; X11 is F, Y, P, W, 
    or D; X14 is S or I; X15 is H, or G; X12 is V, R, or L; 
    T or P; X16 is G, Q, E, L, F, X13 is G or T; X14 is D, S,
    A, S, M, R, K, or Y; X17 is S, T, or A; and X15 is I, S, 
    R, T, G, R, V, D, A, H, E, K, L, P, or D 
    C, F, or Y; X18 is W, S, or F;  (SEQ ID NO: 314)
    and X19 is L or I
    (SEQ ID NO: 316)
    P4G4 EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    ASQSVSSSYLA WYQQKPGQAP ASGFTFS SYAMSWVRQAPGKGLE
    RLLIY GASSRAY GIPDRFSGSGS WVSAISASGGSTYYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YGSPPLFT FGQGTKVEIK  YYCAR LSWSGAFDN WGQGTLVT
    (SEQ ID NO: 80) VSS (SEQ ID NO: 363)
    P1A11 EIVLTQSPGTLSLSPGERATLSC R EVQLLESGGGLVQPGGSLRLSCA
    ASQNVSSSYLA WYQQKPGQAP ASGFTFR SYAMSWVRQAPGKGLE
    RLLIY GASYRAT GIPDRFSGSGS WVSAISGSGGSTFYADSVKGRFTI
    GTDFTLTISRLEPEDFAVYYC QH SRDNSKNTLYLQMNSLRAEDTAV
    YGSPPSFT FGQGTKVEIK  YYCAT VGTSGAFGI WGQGTLVTV
    (SEQ ID NO: 364) SS (SEQ ID NO: 365)
  • In Table 1, the underlined sequences are CDR sequences according to Kabat and in bold according to Chothia, except for the following heavy chain CDR2 sequences, in which the Chothia CDR sequences are underlined and the Kabat CDR sequences are in bold:
      • P5A2_VHVL, A02_Rd4_0.6 nM_C06, A02_Rd4_0.6 nM_C09
      • A02_Rd4_6 nM_C16, A02_Rd4_6 nM_C03, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C26
      • A02_Rd4_6 nM_C25, A02_Rd4_6 nM_C22, A02_Rd4_6 nM_C19, A02_Rd4_0.6 nM_C03
      • A02_Rd4_6 nM_C07, A02_Rd4_6 nM_C23, A02_Rd4_0.6 nM_C18, A02_Rd4_6 nM_C10
      • A02_Rd4_6 nM_C05, A02_Rd4_0.6 nM_C10, A02_Rd4_6 nM_C04,
      • A02_Rd4_0.6 nM_C26
      • A02_Rd4_0.6 nM_C13, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C08, P5C1_VHVL,
      • C01_Rd4_6 nM_C24, C01_Rd4_6 nM_C26, C01_Rd4_6 nM_C10, C01_Rd4_0.6 nM_C27
      • C01_Rd4_6 nM_C20, C01_Rd4_6 nM_C12, C01_Rd4_0.6 nM_C16,
      • C01_Rd4_0.6 nM_C09
      • C01_Rd4_6 nM_C09, C01_Rd4_0.6 nM_C03, C01_Rd4_0.6 nM_C06,
      • C01_Rd4_6 nM_C04
      • COMBO_Rd4_0.6 nM_C22, COMBO_Rd4_6 nM_C21, COMBO_Rd4_6 nM_C10,
      • COMBO_Rd4_0.6 nM_C04, COMBO_Rd4_6 nM_C25, COMBO_Rd4_0.6 nM_C21,
      • COMBO_Rd4_6 nM_C11, COMBO_Rd4_0.6 nM_C20, COMBO_Rd4_6 nM_C09,
      • COMBO_Rd4_6 nM_C08, COMBO_Rd4_0.6 nM_C19, COMBO_Rd4_0.6 nM_C02,
      • COMBO_Rd4_0.6 nM_C23, COMBO_Rd4_0.6 nM_C29, COMBO_Rd4_0.6 nM_C09,
      • COMBO_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C30, COMBO_Rd4_0.6 nM_C14,
      • COMBO_Rd4_6 nM_007, COMBO_Rd4_6 nM_002, COMBO_Rd4_0.6 nM_005,
      • COMBO_Rd4_0.6 nM_017, COMBO_Rd4_6 nM_C22, and COMBO_Rd4_0.6 nM_C11.
  • The invention also provides CDR portions of antibodies to BCMA (including Chothia, Kabat CDRs, and CDR contact regions). Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed “combined CRs” or “extended CDRs”). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof. Table 2 provides examples of CDR sequences provided herein.
  • TABLE 2
    Heavy Chain
    mAb CDRH1 CDRH2 CDRH3
    P6E01 SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    P6E01/P6E01; GFTFGSYAMT (SEQ
    L1.LGF/ ID NO: 131)
    L3.KW/P6E01; (extended)
    L1.LGF/L3.
    NY/P6E01;
    L1.GDF/L3.
    NY/P6E01;
    L3.KW/P6E01;
    L3.PY/P6E01;
    L3.NY/P6E01;
    L3.PY/L1.
    PS/P6E01;
    L3.PY/L1.
    AH/P6E01;
    L3.PY/L1.
    FF/P6E01;
    L3.PY/L1.
    PH/P6E01;
    L3.PY/L3.
    KY/P6E01;
    L3.PY/L3.
    KF/P6E01;
    and
    L3.PY/P6E01.
    H3.AQ SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIAAQMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 135)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    P6E01/H3.AQ; GFTFGSYAMT (SEQ
    L1.LGF/L3. ID NO: 131)
    KW/H3.AQ; (extended)
    L1.LGF/L3.
    PY/H3.AQ;
    L1.LGF/L3.
    NY/H3.AQ;
    L1.GDF/L3.
    KW/H3.AQ;
    L1.GDF/L3.
    PY/H3.AQ;
    L1.GDF/L3.
    NY/H3.AQ;
    L3.PY/H3.
    AQ;
    L3.PY/L1.
    PS/H3.AQ;
    L3.PY/L1.
    AH/H3.AQ;
    L3.PY/L1.
    FF/H3.AQ;
    L3.PY/L1.
    PH/H3.AQ;
    and
    L3.PY/L3.
    KF/H3.AQ.
    H3.AL SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIAALMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 136)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L1.LGF/L3. GFTFGSYAMT (SEQ
    KW/H3.AL; ID NO: 131)
    L1.LGF/L3. (extended)
    NY/H3.AL;
    and
    L1.GDF/L3.
    NY/H3.AL.
    H3.AP SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIAAPMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 137)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L1.LGF/L3. GFTFGSYAMT (SEQ
    KW/H3.AP; ID NO: 131)
    L1.LGF/L3. (extended)
    PY/H3.AP;
    L1.LGF/
    L3NY/H3.AP;
    L1.GDF/L3.
    KW/H3.AP;
    and
    L1.GDF/
    L3NY/H3.AP.
    H2.QR SYAMT (SEQ ID NO: AISGSGGNTFYADQRKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 138) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H2. GFTFGSYAMT (SEQ
    QR; ID NO: 131)
    L3.PY/L1. (extended)
    PS/H2.QR;
    L3.PY/L1.
    AH/H2.QR;
    L3.PY/L1.
    FF/H2.QR;
    L3.PY/L1.
    PH/H2.QR;
    and
    L3.PY/L3.
    KY/H2.QR.
    H2.DY SYAMT (SEQ ID NO: AIDYSGGNTFYADSVKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 139) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID DYSGGN (SEQ ID NO: 140)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H2.DY; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    PS/H2.DY; (extended)
    L3.PY/L1.
    AH/H2.DY;
    L3.PY/L1.
    FF/H2.DY;
    L3.PY/L3.
    KY/H2.DY;
    and
    L3.PY/L3.
    KF/H2.DY.
    H2.YQ SYAMT (SEQ ID NO: AISYQGGNTFYADSVKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 141) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID SYQGGN (SEQ ID NO: 142)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H2.YQ; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    PS/H2.YQ; (extended)
    L3.PY/L1.
    AH/H2.YQ;
    L3.PY/L1.
    FF/H2.YQ;
    L3.PY/L3.
    KY/H2.YQ;
    and
    L3.PY/L3.
    KF/H2.YQ.
    H2.LT SYAMT (SEQ ID NO: AISLTGGNTFYADSVKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 143) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID SLTGGN (SEQ ID NO: 144)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H2.LT; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    PS/H2.LT; (extended)
    L3.PY/L1.
    AH/H2.LT;
    L3.PY/L1.
    FF/H2.LT;
    L3.PY/L3.
    KY/H2.LT;
    and
    L3.PY/L3.
    KF/H2.LT.
    H2.HA SYAMT (SEQ ID NO: AISHAGGNTFYADSVKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 145) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID SHAGGN (SEQ ID NO: 146)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H2.HA; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    AH/H2.HA; (extended)
    L3.PY/L1.
    FF/H2.HA;
    L3.PY/L1.
    PH/H2.HA;
    and
    L3.PY/L3.
    KY/H2.HA.
    H2.QL SYAMT (SEQ ID NO: AISGSGGNTFYADQLKG VSPIASGMDY
    For the 129) (Kabat); (SEQ ID NO: 147) (Kabat) (SEQ ID NO: 134)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H2.QL; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    PS/H2.QL; (extended)
    L3.PY/L1.
    AH/H2.QL;
    L3.PY/L1.
    FF/H2.QL;
    L3.PY/L3.
    KY/H2.QL;
    and
    L3.PY/L3.
    KF/H2.QL.
    H3.YA SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIYAGMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 148)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H3.YA; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    PS/H3.YA; (extended)
    L3.PY/L1.
    AH/H3.YA;
    L3.PY/L1.
    FF/H3.YA;
    L3.PY/L3.
    KY/H3.YA;
    and
    L3.PY/L3.
    KF/H3.YA.
    H3.AE SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIAAEMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 149)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H3.AE; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    AH/H3.AE; (extended)
    L3.PY/L1.
    FF/H3.AE;
    L3.PY/L1.
    H/H3.AE;
    Pand
    L3.PY/L3.
    KF/H3.AE.
    H3.TAQ SYAMT (SEQ ID NO: AISGSGGNTFYADSVKG VSPIAAQMDY
    For the 129) (Kabat); (SEQ ID NO: 132) (Kabat) (SEQ ID NO: 135)
    following GFTFGSY (SEQ ID SGSGGN (SEQ ID NO: 133)
    mAbs: NO: 130) (Chothia); (Chothia)
    L3.PY/H3.TAQ; GFTFGSYAMT (SEQ
    L3.PY/L1. ID NO: 131)
    PS/H3.TAQ; (extended)
    L3.PY/L1.
    AH/H3.TAQ;
    L3.PY/L1.
    FF/H3.TAQ;
    L3.PY/L1.
    PH/H3.TAQ;
    and
    L3.PY/L3.
    KF/H3.TAQ.
    P5A2_VH SYAMN (SEQ ID NO: AISDSGGSTYYADSVKG YWPMDI (SEQ ID
    VL and 150) (Kabat); (SEQ ID NO: 153) (Kabat) NO: 155)
    A02_Rd4_ GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    6nM_C03 NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    COMBO SYPMS (SEQ ID NO: AIGGSGGSLPYADSVKG YWPMDI (SEQ ID
    Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 158) (Kabat) NO: 155)
    C17; GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    COMBO NO: 151) (Chothia); (Chothia)
    Rd4_0.6 nM_ GFTFSSYPMS (SEQ
    C14; ID NO: 157)
    COMBO (extended)
    Rd4_0.6 nM_
    C29;
    and
    COMBO_
    Rd4_0.6 nM_
    C09
    C01_Rd4_ SYPMS (SEQ ID NO: AIGGSGGSLPYADSVKG YWPMDS (SEQ
    6 nM_C04; 156) (Kabat); (SEQ ID NO: 158) (Kabat) ID NO: 161)
    C01_Rd4_ GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    0.6 nM NO: 151) (Chothia); (Chothia)
    C03; GFTFSSYPMS (SEQ
    C01_Rd4_ ID NO: 157)
    0.6 nM (extended)
    C06;
    COMBO_
    Rd4_0.6 nM_
    C02;
    COMBO_
    Rd4_6 nM_
    C21;
    C01_Rd4_
    6nM_C26;
    COMBO_
    Rd4_0.6 nM_
    C19;
    C01_Rd4_
    6 nM_C24;
    C01_Rd4_
    6 nM_C20;
    C01_Rd4_
    0.6 nM_
    C09;
    COMBO_
    Rd4_0.6 nM_
    C21;
    C01_Rd4_
    0.6 nM_
    C04_C27;
    C01_Rd4_
    0.6 nM_
    C16;
    C01_Rd4_
    6 nM_C10;
    COMBO_
    Rd4_0.6 nM_
    C20
    P5C1_ SYPMS (SEQ ID NO: AIGGSGGSTYYADSVKG YWPMDS (SEQ
    VHVL and 156) (Kabat); (SEQ ID NO: 162) (Kabat) ID NO: 161)
    COMBO_ GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    Rd4_0.6 nM_ NO: 151) (Chothia); (Chothia)
    C30 GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSAWYADSVKG YWPMSL (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 163) (Kabat) ID NO: 164)
    C06 GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSAWYADSVKG YWPMSL (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 163) (Kabat) ID NO: 164)
    C09 GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDFGGSTYYADSVKG YWPMDI (SEQ ID
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 165) (Kabat) NO: 155)
    C16 GFTFSSY (SEQ ID SDFGGS (SEQ ID NO: 166)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AITASGGSTYYADSVKG YWPMSL (SEQ
    6 nM_C01 150) (Kabat); (SEQ ID NO: 167) (Kabat) ID NO: 164)
    GFTFSSY (SEQ ID TASGGS (SEQ ID NO: 168)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSTYYADSVKG YWPMSL (SEQ
    6 nM_C26 150) (Kabat); (SEQ ID NO: 153) (Kabat) ID NO: 164)
    GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSRWYADSVKG YWPMTP (SEQ
    6 nM_C25 150) (Kabat); (SEQ ID NO: 169) (Kabat) ID NO: 170)
    GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AVLDSGGSTYYADSVKG YWPMTP (SEQ
    6 nM_C22 150) (Kabat); (SEQ ID NO: 171) (Kabat) ID NO: 170)
    GFTFSSY (SEQ ID LDSGGS (SEQ ID NO: 172)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSRWYADSVKG YWPMSD (SEQ
    6 nM_C19 150) (Kabat); (SEQ ID NO: 169) (Kabat) ID NO: 173)
    GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSKWYADSVKG YWPMSL (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 174) (Kabat) ID NO: 164)
    C03 GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AIGGSGGSLPYADSVKG(S YWPMDS (SEQ
    6 nM_C07 150) (Kabat); EQ ID NO: 158) (Kabat) ID NO: 161)
    GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSGWYADSVKG YWPMSL (SEQ
    6 nM_C23 150) (Kabat); (SEQ ID NO: 175) ID NO: 164)
    GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AVLDSGGSTYYADSVKG YWPMSL (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 171) (Kabat) ID NO: 164)
    C18 GFTFSSY (SEQ ID LDSGGS (SEQ ID NO: 172)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGSCWYADSVKG YWPMTP (SEQ
    6 nM_C10 150) (Kabat); (SEQ ID NO: 176) (Kabat) ID NO: 170)
    GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AIFASGGSTYYADSVKG YWPMTP (SEQ
    6 nM_C05 150) (Kabat); (SEQ ID NO: 177) (Kabat) ID NO: 170)
    GFTFSSY (SEQ ID FASGGS (SEQ ID NO: 178)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISGWGGSLPYADSVKG YWPMDS (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 304) (Kabat) ID NO: 161)
    C10 GFTFSSY (SEQ ID SGWGGS (SEQ ID NO: 179)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AIMSSGGPLYYADSVKG YWPMAL (SEQ
    6 nM_C04 150) (Kabat); (SEQ ID NO: 180) (Kabat) ID NO: 182)
    GFTFSSY (SEQ ID MSSGGP (SEQ ID NO: 181)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AILMSGGSTYYADSVKG YWPMSL (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 183) (Kabat) ID NO: 164)
    C26 GFTFSSY (SEQ ID LMSGGS (SEQ ID NO: 184)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AISDSGGYRYYADSVKG YWPMSL (SEQ
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 185) (Kabat) ID NO: 164)
    C13 GFTFSSY (SEQ ID SDSGGY (SEQ ID NO: 186)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AILSSGGSTYYADSVKG YWPMDI (SEQ ID
    0.6 nM_ 150) (Kabat); (SEQ ID NO: 187) (Kabat) NO: 155)
    C01 GFTFSSY (SEQ ID LSSGGS (SEQ ID NO: 188)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    A02_Rd4_ SYAMN (SEQ ID NO: AILDSGGSTYYADSVKG YWPMSP (SEQ
    6 nM_C08 150) (Kabat); (SEQ ID NO: 160) (Kabat) ID NO: 189)
    GFTFSSY (SEQ ID LDSGGS (SEQ ID NO: 172)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    C01_Rd4_ SYPMS (SEQ ID NO: AIGGSGGWSYYADSVKG YWPMDS (SEQ
    6 nM_C12 156) (Kabat); (SEQ ID NO: 190) (Kabat) ID NO: 161)
    GFTFSSY (SEQ ID GGSGGW (SEQ ID NO: 191)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    C01_Rd4_ SYPMS (SEQ ID NO: ATVGSGGSIGYADSVKG YWPMDS (SEQ
    6 nM_C09 156) (Kabat); (SEQ ID NO: 192) (Kabat) ID NO: 161)
    GFTFSSY (SEQ ID VGSGGS (SEQ ID NO: 193)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYAMN (SEQ ID NO: AISDSGGSRWYADSVKG YWPMDI (SEQ ID
    Rd4_0.6 nM_ 150) (Kabat); (SEQ ID NO: 169) (Kabat) NO: 155)
    C22 GFTFSSY (SEQ ID SDSGGS (SEQ ID NO: 154)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIGGSGGSIHYADSVKG YWPMDS (SEQ
    Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 194) (Kabat) ID NO: 161)
    C10 GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AHIGSGGSTYYADSVKG YWPMDS (SEQ
    Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 195) (Kabat) ID NO: 161)
    C04 GFTFSSY (SEQ ID IGSGGS (SEQ ID NO: 196)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIGGSGGSTYYADSVKG YWPMDP (SEQ
    Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 162) (Kabat) ID NO: 197)
    C25 GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIGGSGGSLPYADSVKG YWPMDS (SEQ
    Rd4_6 nM_ 156) (Kabat); (SEQ ID NO: 158) (Kabat) ID NO: 161)
    C21 GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIGGSGGSLGYADSVKG YWPMDS (SEQ
    Rd4_6 nM_ 156) (Kabat); (SEQ ID NO: 198) (Kabat) ID NO: 161)
    C11 GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIFASGGSTYYADSVKG YWPMDS (SEQ
    Rd4_6 nM_ 156) (Kabat); (SEQ ID NO: 177) (Kabat) ID NO: 161)
    C09 GFTFSSY (SEQ ID FASGGS (SEQ ID NO: 178)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIGGSGTWTYYADSVKG YWPMDS (SEQ
    Rd4_6 nM_ 156) (Kabat); (SEQ ID NO: 199) (Kabat) ID NO: 161)
    C08 GFTFSSY (SEQ ID GGSGTW (SEQ ID NO: 200)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: ALFGSGGSTYYADSVKG YWPMDS (SEQ
    Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 201) (Kabat) ID NO: 161)
    C23 GFTFSSY (SEQ ID FGSGGS
    NO: 151) (Chothia); (SEQ ID NO: 202) (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AALGSGGSTYYADSVKG YWPMDS (SEQ
    Rd4_0.6 nM_ 156) (Kabat); (SEQ ID NO: 203) (Kabat) ID NO: 161)
    C12 GFTFSSY (SEQ ID LGSGGS (SEQ ID NO: 204)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AIGGSGGSLPYADSVKG YWPMAD (SEQ
    Rd4_6 nM_ 156) (Kabat); (SEQ ID NO: 158) (Kabat) ID NO: 205)
    C07 GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    COMBO_ SYAMN (SEQ ID NO: AISDSGGFVYYADSVKG YWPMDS (SEQ
    Rd4_6 nM_ 150) (Kabat); (SEQ ID NO: 206) (Kabat) ID NO: 161)
    C02 GFTFSSY (SEQ ID SDSGGF (SEQ ID NO: 207)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    COMBO_ SYAMN (SEQ ID NO: AIGGSGGSTYYADSVKG YWPMSL (SEQ
    Rd4_6 nM_ 150) (Kabat); (SEQ ID NO: 162) (Kabat) ID NO: 164)
    C05 GFTFSSY (SEQ ID GGSGGS (SEQ ID NO: 159)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    COMBO_ SYAMN (SEQ ID NO: ACLDSGGSTYYADSVKG YWPMDS (SEQ
    Rd4_6 nM_ 150) (Kabat); (SEQ ID NO: 208) (Kabat) ID NO: 161)
    C22 GFTFSSY (SEQ ID LDSGGS (SEQ ID NO: 172)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYAMN (SEQ
    ID NO: 152)
    (extended)
    COMBO_ SYPMS (SEQ ID NO: AALGSGGSTYYADSVKG YWPMSL (SEQ
    Rd4_6 nM_ 156) (Kabat); (SEQ ID NO: 203) (Kabat) ID NO: 164)
    C11 GFTFSSY (SEQ ID LGSGGS (SEQ ID NO: 204)
    NO: 151) (Chothia); (Chothia)
    GFTFSSYPMS (SEQ
    ID NO: 157)
    (extended)
    Heavy SYX1MX2, wherein X1 AX1X2X3X4GX5X6X7X8YADX9 VSPIX1X2X3MDY,
    chain is A or P; and X2 is X10KG, wherein X1 is I, V, T, wherein X1 is A or
    consensus T, N, or S (Kabat) H, L, A, or C; X2 is S, D, G, T, Y; X2 is A or S; and
    (SEQ ID NO: 301) I, L, F, M, or V; X3 is G, Y, L, X3 is G, Q, L, P, or
    GFTFX1SY, wherein H, D, A, S, or M; X4 is S, Q, T, E (SEQ ID NO: 307)
    X1 is G or S (Chothia) A, F, or W; X5 is G or T; X6 is YWPMX1X2,
    (SEQ ID NO: 302) N, S, P, Y, W, or F; X7 is S, T, wherein X1 is D, S,
    GFTFX1SYX2MX3, I, L, A, R, V, K, G, or C; X8 is T, or A; and X2 is I,
    wherein X1 is G or S, F, Y, P, W, H, or G; X9 is V, R, S, L, P, or D (SEQ
    X2 is A or P; and X3 or L; and X10 is G or T (Kabat) ID NO: 308)
    is T, N, or S (SEQ ID (SEQ ID NO: 305)
    NO: 303) (extended) X1X2X3X4X5X6, wherein X1 is
    S, V, I, D, G, T, L, F, or M; X2
    is G, Y, L, H, D, A, S, or M; X3
    is S, G, F, or W; X4 is G or S;
    X5 is G or T; and X6 is N, S,
    P, Y, or W (Chothia) (SEQ ID
    NO: 306)
    P4G4 SYAMS (SEQ ID NO: SASGGS (SEQ ID NO: 368) LSWSGAFDN
    366) (Kabat); (Chothia) (SEQ ID NO: 370)
    GFTFSSY (SEQ ID AISASGGSTYYADSVKG
    NO: 151) (Chothia); (SEQ ID NO: 369) (Kabat)
    GFTFSSYAMS (SEQ
    ID NO: 367)
    (extended)
    P1A11 SYAMS (SEQ ID NO: SGSGGS (SEQ ID NO: 359) VGTSGAFGI
    366) (Kabat); (Chothia) (SEQ ID NO: 361)
    GFTFRSY (SEQ ID AISGSGGSTFYADSVKG
    NO: 371) (SEQ ID NO: 360) (Kabat)
    GFTFRSYAMS (SEQ
    ID NO: 372)
    Light Chain
    mAb CDRL1 CDRL2 CDRL3
    P6E01 RASQSVSSSYLA GASSRAT (SEQ ID NO: QHYGSPPSFT
    For the (SEQ ID NO: 209) 210) (SEQ ID NO: 211)
    following
    mAbs:
    P6E01/
    P6E01; and
    P6E01/H3.AQ.
    L1.LGF/ RASQSLGSFYLA GASSRAT (SEQ ID NO: KHYGWPPSFT
    L3.KW (SEQ ID NO: 212) 210) (SEQ ID NO: 213)
    For the
    following
    mAbs:
    L1.LGF/
    L3.KW/P6E01;
    L1.LGF/
    L3.KW/H3.AL;
    L1.LGF/
    L3.KW/H3.
    AP; and
    L1.LGF/
    L3KW/H3.AQ
    L1.LGF/ RASQSLGSFYLA GASSRAT (SEQ ID NO: QHYNYPPSFT
    L3.NY (SEQ ID NO: 212) 210) (SEQ ID NO: 214)
    For the
    following
    mAbs:
    L1.LGF/
    L3.NY/P6E01;
    L1.LGF/
    L3.NY/H3.AL;
    L1.LGF/
    L3.NY/H3.AP;
    and
    L1.LGF/
    L3.NY/H3AQ
    L1.GDF/ RASQSVGDFYLA GASSRAT (SEQ ID NO: QHYNYPPSFT
    L3.NY (SEQ ID NO: 215) 210) (SEQ ID NO: 214)
    For the
    following
    mAbs:
    L1.GDF/
    L3.NY/P6E01;
    L1.GDF/
    L3.NY/H3.AL;
    L1.GDF/
    L3.NY/H3.
    AP; and
    L1.GDF/
    L3.NY/H3.AQ
    L1.LGF/ RASQSLGSFYLA GASSRAT (SEQ ID NO: QHYPYPPSFT
    L3.PY (SEQ ID NO: 212) 210) (SEQ ID NO: 216)
    For the
    following
    mAbs:
    L1.LGF/
    L3.PY/H3.
    AP; and
    L1.LGF/
    L3.PY/H3.
    AQ
    L1.GDF/ RASQSVGDFYLA GASSRAT (SEQ ID NO: KHYGWPPSFT
    L3.KW (SEQ ID NO: 215) 210) (SEQ ID NO: 213)
    For the
    following
    mAbs:
    L1.GDF/
    L3.KW/H3.AL;
    L1.GDF/
    L3.KW/
    H3.AP; and
    L1.GDF/
    L3.KW/
    H3.AQ
    L1.GDF/ RASQSVGDFYLA GASSRAT (SEQ ID NO: QHYPYPPSFT
    L3.PY/ (SEQ ID NO: 215) 210) (SEQ ID NO: 216)
    H3.AQ
    L3.KW/ RASQSVSSSYLA GASSRAT (SEQ ID NO: KHYGWPPSFT
    P6E01 (SEQ ID NO: 209) 210) (SEQ ID NO: 213)
    L3.PY RASQSVSSSYLA GASSRAT (SEQ ID NO: QHYPYPPSFT
    For the (SEQ ID NO: 209) 210) (SEQ ID NO: 216)
    following
    mAbs:
    L3.PY/
    P6E01;
    L3.PY/H2.
    QR;
    L3.PY/H2.
    DY;
    L3.PY/H2.
    YQ;
    L3.PY/H2.
    LT;
    L3.PY/H2.
    HA;
    L3.PY/H2.
    QL;
    L3.PY/H3.
    YA
    L3.PY/H3.
    AE
    L3.PY/H3.
    AQ;
    L3.PY/H3.
    TAQ
    L3.NY/ RASQSVSSSYLA GASSRAT (SEQ ID NO: QHYNYPPSFT
    P6E01 (SEQ ID NO: 209) 210) (SEQ ID NO: 214)
    L3.PY/ RASQSVSSSYPS GASSRAT (SEQ ID NO: QHYPYPPSFT
    L1.PS (SEQ ID NO: 217) 210) (SEQ ID NO: 216)
    For the
    following
    mAbs:
    L3.PY/L1.
    PS/
    P6E01;
    L3.PY/L1.
    PS/H2.QR;
    L3.PY/L1.
    PS/H2.DY;
    L3.PY/L1.
    PS/H2.YQ;
    L3.PY/L1.
    PS/H2.LT;
    L3.PY/L1.
    PS/H2.HA;
    L3.PY/L1.
    PS/H2.QL;
    L3.PY/L1.
    PS/H3.YA;
    L3.PY/L1.
    PS/H3.AE;
    L3.PY/L1.
    PS/H3.AQ;
    L3.PY/L1.
    PS/H3.TAQ
    L3.PY/ RASQSVSAHYLA GASSRAT (SEQ ID NO: QHYPYPPSFT
    L1.AH (SEQ ID NO: 218) 210) (SEQ ID NO: 216)
    For the
    following
    mAbs:
    L3.PY/L1.
    AH/P6E01;
    L3.PY/L1.
    AH/H2.QR;
    L3.PY/L1.
    AH/H2.DY;
    L3.PY/L1.
    AH/H2.YQ;
    L3.PY/L1.
    AH/H2.LT;
    L3.PY/L1.
    AH/H2.HA;
    L3.PY/L1.
    AH/H2.QL;
    L3.PY/L1.
    AH/H3.YA;
    L3.PY/L1.
    AH/H3.AE;
    L3.PY/L1.
    AH/H3.AQ;
    L3.PY/L1.
    AH/H3.TAQ
    L3.PY/ RASQSVSSFFLA GASSRAT (SEQ ID NO: QHYPYPPSFT
    L1.FF (SEQ ID NO: 219) 210) (SEQ ID NO: 216)
    For the
    following
    mAbs:
    L3.PY/L1.
    FF/P6E01;
    L3.PY/L1.
    FF/H2.QR;
    L3.PY/L1.
    FF/H2.DY;
    L3.PY/L1.
    FF/H2.YQ;
    L3.PY/L1.
    FF/H2.LT;
    L3.PY/L1.
    FF/H2.HA;
    L3.PY/L1.
    FF/H2.QL;
    L3.PY/L1.
    FF/H3.YA;
    L3.PY/L1.
    FF/H3.AE;
    L3.PY/L1.
    FF/H3.AQ;
    and
    L3.PY/L1.
    FF/H3.TAQ
    L3.PY/ RASQSVSPHYLA GASSRAT (SEQ ID NO: QHYPYPPSFT
    L1.PH (SEQ ID NO: 219) 210) (SEQ ID NO: 216)
    For the
    following
    mAbs:
    L3.PY/L1.
    PH/P6E01;
    L3.PY/L1.
    PH/H2.QR;
    L3.PY/L1.
    PH/H2.HA;
    L3.PY/L1.
    PH/H3.AE;
    L3.PY/L1.
    PH/H3.AQ;
    and
    L3.PY/L1.
    PH/H3.TAQ
    L3.PY/ RASQSVSSSYLA GASSRAT (SEQ ID NO: KYYPYPPSFT
    L3.KY (SEQ ID NO: 209) 210) (SEQ ID NO: 220)
    For the
    following
    mAbs:
    L3.PY/L3.
    KY/P6E01;
    L3.PY/L3.
    KY/H2.QR;
    L3.PY/L3.
    KY/H2.DY;
    L3.PY/L3.
    KY/H2.YQ;
    L3.PY/L3.
    KY/H2.LT;
    L3.PY/L3.
    KY/H2.HA;
    L3.PY/L3.
    KY/H2.QL;
    L3.PY/L3.
    KY/H3.YA;
    and
    L3.PY/L3.
    KY/H3.TAQ
    L3.PY/ RASQSVSSSYLA GASSRAT (SEQ ID NO: KFYPYPPSFT (SEQ
    L3.KF (SEQ ID NO: 209) 210) ID NO: 220)
    For the
    following
    mAbs:
    L3.PY/L3.
    KF/H2.DY;
    L3.PY/L3.
    KF/H2.YQ;
    L3.PY/L3.
    KF/H2.LT;
    L3.PY/L3.
    KF/H2.QL;
    L3.PY/L3.
    KF/H3.YA;
    L3.PY/L3.
    KF/H3.AE;
    L3.PY/L3.
    KF/H3.AQ;
    and
    L3.PY/L3.
    KF/H3.TAQ
    P5A2_ RASQSVSSSYLA DASIRAT QQYGSWPLT (SEQ
    VHVL (SEQ ID NO: 209) (SEQ ID NO: 221) ID NO: 222)
    A02_Rd4_ RASQSVSVIYLA DASIRAT QQYQRWPLT
    0.6 nM_ (SEQ ID NO: 223) (SEQ ID NO: 221) (SEQ ID NO: 224)
    C06
    A02_Rd4_ RASQSVSSSYLA DASIRAT QQYQSWPLT
    0.6 nM_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 225)
    C09;
    COMBO_
    Rd_0.6
    nM_C29;
    and
    COMBO_
    Rd4_
    0.6 nM_C21
    A02_Rd4_ RASQSVSDIYLA DASIRAT QQYQTWPLT (SEQ
    6 nM_ (SEQ ID NO: 226) (SEQ ID NO: 221) ID NO: 227)
    C16
    A02_Rd4_ RASQSVSNIYLA DASIRAT QQYQGWPLT (SEQ
    6 nM_ (SEQ ID NO: 228) (SEQ ID NO: 221) ID NO: 229)
    C03
    A02_Rd4_ RASQSVSAYYLA DASIRAT QQYERWPLT
    6 nM_ (SEQ ID NO: 230) (SEQ ID NO: 221) (SEQ ID NO: 231)
    C01
    A02_Rd4_ RASQSVSSIYLA DASIRAT QQYQVWPLT
    6 nM_ (SEQ ID NO: 232) (SEQ ID NO: 221) (SEQ ID NO: 233)
    C26
    A02_Rd4_ RASQSVSSSYLA DASIRAT QQYLDWPLT
    6 nM_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 234)
    C25
    A02_Rd4_ RASQSVSSSYLA DASIRAT QQYQVWPLT
    6 nM_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 233)
    C22
    A02_Rd4_ RASQSVSVIYLA DASIRAT QQYLAWPLT
    6 nM_ (SEQ ID NO: 223) (SEQ ID NO: 221) (SEQ ID NO: 236)
    C19
    A02_Rd4_ RASQSVSSSYLA DASIRAT QQYFTWPLT
    0.6 nM_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 237)
    C03
    A02_Rd4_ RASQSVSPYYLA DASIRAT QQYERWPLT
    6 nM_ (SEQ ID NO: 238) (SEQ ID NO: 221) (SEQ ID NO: 231)
    C07
    A02_Rd4_ RASQSVSVEYLA DASIRAT QQYARWPLT
    6 nM_ (SEQ ID NO: 239) (SEQ ID NO: 221) (SEQ ID NO: 240)
    C23
    A02_Rd4_ RASQSVSEIYLA DASIRAT QQYFGWPLT
    0.6 nM_ (SEQ ID NO: 241) (SEQ ID NO: 221) (SEQ ID NO: 242)
    C18
    A02_Rd4_ RASQSVEMSYLA DASIRAT QQYAHWPLT
    6 nM_ (SEQ ID NO: 243) (SEQ ID NO: 221) (SEQ ID NO: 244)
    C10
    A02_Rd4_ RASQSVSSSYLA DASIRAT QQYQRWPLT
    6 nM_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 224)
    C05
    A02_Rd4_ RASQSVSAQYLA DASIRAT QQYQRWPLT
    0.6 nM_ (SEQ ID NO: 245) (SEQ ID NO: 221) (SEQ ID NO: 224)
    C10
    A02_Rd4_ RASQSVSAIYLA DASIRAT QQYQVWPLT
    6 nM_ (SEQ ID NO: 235) (SEQ ID NO: 221) (SEQ ID NO: 233)
    C04
    A02_Rd4_ GPSQSVSSSYLA DASIRAT QQYQSWPLT
    0.6 nM_ (SEQ ID NO: 246) (SEQ ID NO: 221) (SEQ ID NO: 225)
    C26
    A02_Rd4_ RASQSVSSSYWA DASIRAT QQYESWPLT
    0.6 nM_ (SEQ ID NO: 247) (SEQ ID NO: 221) (SEQ ID NO: 248)
    C13
    A02_Rd4_ RGGQSVSSSYLA DASIRAT QQYQSWPLT
    0.6 nM_ (SEQ ID NO: 249) (SEQ ID NO: 221) (SEQ ID NO: 225)
    C01
    A02_Rd4_ RASQSVSFIYLA DASIRAT QQYGSWPLT (SEQ
    6 nM_ (SEQ ID NO: 250) (SEQ ID NO: 221) ID NO: 222)
    C08
    P5C1_ RASQSVSSTYLA DASSRAP QQYSTSPLT
    VHVL (SEQ ID NO: 251) (SEQ ID NO: 252) (SEQ ID NO: 253)
    C01_Rd4_ RASQSVSPEYLA DASSRAP QQYSVWPLT
    6 nM_ (SEQ ID NO: 254) (SEQ ID NO: 252) (SEQ ID NO: 255)
    C24
    C01_Rd4_ RASQSVSAIYLA DASSRAP QQYSAWPLT
    6 nM_ (SEQ ID NO: 235) (SEQ ID NO: 252) (SEQ ID NO: 256)
    C26
    C01 Rd4_ RASQSVSSVYLA DASSRAP QQYSTWPLT
    6 nM_ (SEQ ID NO: 257) (SEQ ID NO: 252) (SEQ ID NO: 258)
    C10
    C01_Rd4_ RASQSVSSTYLA DASSRAP QQYSRWPLT
    0.6 nM_ (SEQ ID NO: 251) (SEQ ID NO: 252) (SEQ ID NO: 259)
    C27
    C01_Rd4_ RASQSVSPIYLA DASSRAP QQYSAFPLT
    6 nM_ (SEQ ID NO: 260) (SEQ ID NO: 252) (SEQ ID NO: 261)
    C20
    C01_Rd4_ WLSQSVSSTYLA DASSRAP QQYSEWPLT
    6 nM_ (SEQ ID NO: 262) (SEQ ID NO: 252) (SEQ ID NO: 263)
    C12
    C01_Rd4_ RASQSVSSTYLA DASSRAP QQYSSWPLT
    0.6 nM_ (SEQ ID NO: 251) (SEQ ID NO: 252) (SEQ ID NO: 264)
    C16
    C01_Rd4_ RASQSVSSIFLA DASSRAP QQYSAWPLT
    0.6 nM_ (SEQ ID NO: 265) (SEQ ID NO: 252) (SEQ ID NO: 256)
    C09
    C01_Rd4_ ACSQSVSSTYLA DASSRAP QQYSAWPLT
    6 nM_ (SEQ ID NO: 266) (SEQ ID NO: 252) (SEQ ID NO: 256)
    C09
    C01_Rd4_ RASCDVSSTYLA DASSRAP QQYMRSPLT
    0.6 nM_ (SEQ ID NO: 267) (SEQ ID NO: 252) (SEQ ID NO: 268)
    C03
    C01_Rd4_ RASEAVPSTYLA DASSRAP QQYSAFPLT
    0.6 nM_ (SEQ ID NO: 269) (SEQ ID NO: 252) (SEQ ID NO: 261)
    C06
    C01_Rd4_ CSSQSVSSTYLA DASSRAP QQYSAFPLT
    0.6 nM_ (SEQ ID NO: 270) (SEQ ID NO: 252) (SEQ ID NO: 261)
    C04
    COMBO_ RASVRVSSTYLA DASIRAT QQYMKWPLT
    Rd4_ (SEQ ID NO: 271) (SEQ ID NO: 221) (SEQ ID NO: 272)
    0.6 nM_C22
    COMBO_ RASQSVSAAYLA DASIRAT QQYMCWPLT
    Rd4_ (SEQ ID NO: 273) (SEQ ID NO: 221) (SEQ ID NO: 274)
    6 nM_C21
    COMBO_ RASQSVSSSYWG DASIRAT QQYQCWPLT
    Rd4_ (SEQ ID NO: 275) (SEQ ID NO: 221) (SEQ ID NO: 276)
    6 nM_C10
    COMBO_ RASQSVSSTYLA DASIRAT QQYQSWPLT
    Rd4_ (SEQ ID NO: 251) (SEQ ID NO: 221) (SEQ ID NO: 225)
    0.6 nM_C04
    COMBO_ RASQSVSSPYLA DASIRAT QQYQSWPLT
    Rd4_ (SEQ ID NO: 277) (SEQ ID NO: 221) (SEQ ID NO: 225)
    6 nM_C25
    COMBO_ RASQSVSPIYLA DASIRAT QQYKAWPLT
    Rd4_ (SEQ ID NO: 260) (SEQ ID NO: 221) (SEQ ID NO: 278)
    6 nM_C11
    COMBO_ RASQSVSYLYLA DASIRAT QQYMEWPLT
    Rd4_ (SEQ ID NO: 279) (SEQ ID NO: 221) (SEQ ID NO: 280)
    0.6 nM_C20
    COMBO_ RASQSVSAQYLA DASIRAT QQYQAWPLT
    Rd4_ (SEQ ID NO: 245) (SEQ ID NO: 221) (SEQ ID NO: 281)
    6 nM_C09
    COMBO_ RASQSVSSSYLA DASIRAT QQYQKWPLT
    Rd4_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 282)
    6 nM_C08
    COMBO_ RASQSVSAVYLA DASIRAT QQYRAWPLT
    Rd4_ (SEQ ID NO: 283) (SEQ ID NO: 221) (SEQ ID NO: 284)
    0.6 nM_C19
    COMBO_ RASIAVSSTYLA DASIRAT QQYMVWPLT
    Rd4_ (SEQ ID NO: 285) (SEQ ID NO: 221) (SEQ ID NO: 286)
    0.6 nM_C02
    COMBO_ RPRQSVSSSYLA DASIRAT QQYQDWPLT
    Rd4_ (SEQ ID NO: 287) (SEQ ID NO: 221) (SEQ ID NO: 288)
    0.6 nM_
    C23
    COMBO_ RASQSVSSTYLA DASIRAT QQYQEWPLT
    Rd4_ (SEQ ID NO: 251) (SEQ ID NO: 221) (SEQ ID NO: 289)
    0.6 nM_
    C09
    COMBO_ RASQSVSASYLA DASIRAT QQYMSWPLT
    Rd4_ (SEQ ID NO: 290) (SEQ ID NO: 221) (SEQ ID NO: 291)
    6 nM_C12
    COMBO_ RASQSVSYMYLA DASIRAT QQYKSWPLT
    Rd4_ (SEQ ID NO: 292) (SEQ ID NO: 221) (SEQ ID NO: 293)
    0.6 nM_
    C30
    COMBO_ RASQSVSAIYLA DASIRAT QQYYGWPLT
    Rd4_ (SEQ ID NO: 235) (SEQ ID NO: 221) (SEQ ID NO: 294)
    0.6 nM_
    C14
    COMBO_ RASQPISSSYLA DASIRAT QQYQGWPLT
    Rd4_ (SEQ ID NO: 295) (SEQ ID NO: 221) (SEQ ID NO: 229)
    6 nM_C07
    COMBO_ RASQSVSSSYLA DASIRAT QQYEFWPLT
    Rd4_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 296)
    6 nM_C02
    COMBO_ RASQSVSSTYLA DASIRAT QQYMSWPLT
    Rd4_ (SEQ ID NO: 251) (SEQ ID NO: 221) (SEQ ID NO: 291)
    0.6 nM_
    C05
    COMBO_ RASQGISSTYLA DASIRAT QQYAYWPLT
    Rd4_ (SEQ ID NO: 297) (SEQ ID NO: 221) (SEQ ID NO: 298)
    0.6 nM_
    C17
    COMBO_ RASQSVSSSYLA DASIRAT QQYQGWPLT
    Rd4_ (SEQ ID NO: 209) (SEQ ID NO: 221) (SEQ ID NO: 229)
    6 nM_C22
    COMBO_ RASQSVSVRYLA DASIRAT QQYGSWPIT
    Rd4_ (SEQ ID NO: 299) (SEQ ID NO: 221) (SEQ ID NO: 300)
    0.6 nM_
    C11
    Light X1X2X3X4X5X6X7X8X9 X1ASX2RAX3, wherein X1 X1X2YX3X4PPSFT,
    chain X10X11X12, wherein X1 is G or D; X2 is S or I; and wherein X1 is Q or K;
    consensus is R, G, W, A, or C; X3 is T or P (SEQ ID NO: X2 is H or Y; X3 is G,
    X2 is A, P, G, L, C, 310) N, or P; and X4 is S,
    or S; X3 is S, G, or W, or Y (SEQ ID NO:
    R; X4 is Q, C, E, V, 311)
    or I; X5 is S, P, QQYX1X2X3PX4T,
    G, A, R, or D; X6 is wherein X1 is G, Q,
    V, G, I, or L; X7 is E, L, F, A, S, M, K,
    S, E, D, P, or G; X8 R, or Y; X2 is S, R,
    is S, P, F, A, M, E, T, G, V, F, Y, D, A,
    V, N, D, or Y; X9 is H, V, E, K, or C; X3
    I, T, V, E, F S, A, M, is W, F, or
    Q, Y, H, or R; X10 is S; and X4 is L or I
    Y or F; X11 is L, W, (SEQ ID NO: 312)
    or P; and X12 is A, S,
    or G (SEQ ID NO: 309)
    P4G4 RASQSVSSSYLA GASSRAY (SEQ ID NO: QHYGSPPLFT
    (SEQ ID NO: 209) 362) (SEQ ID NO: 499)
    P1A11 RASQNVSSSYLA GASYRAT (SEQ ID NO: QHYGSPPSFT
    (SEQ ID NO: 500) 501) (SEQ ID NO: 211)
  • In some embodiments, the present invention provides an antibody that binds to BCMA and competes with the antibody as described herein, including P6E01/P6E01, P6E01/H3.AQ, Li.LGF/L3.KW/P6E01; Li.LGF/L3.NY/P6E01, Li.GDF/L3.NY/P6E01, Li.LGF/L3.KW/H3.AL, Li.LGF/L3.KW/H3.AP, Li.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.NY/H3.AL, L1.GDF/L3.NY/H3.AP, L1.GDF/L3.NY/H3.AQ, L3.KW/P6E01, L3.PY/P6E01, L3.NY/P6E01,
      • L3.PY/L1.PS/P6E01, L3.PY/L1.AH/P6E01, L3.PY/L1.FF/P6E01, L3.PY/L1.PH/P6E01, L3.PY/L3.KY/P6E01, L3.PY/L3.KF/P6E01, L3.PY/H2.QR, L3.PY/H2.DY, L3.PY/H2.YQ, L3.PY/H2.LT, L3.PY/H2.HA, L3.PY/H2.QL, L3.PY/H3.YA, L3.PY/H3.AE, L3.PY/H3.AQ, L3.PY/H3.TAQ, L3.PY/P6E01, L3.PY/L1.PS/H2.QR, L3.PY/L1.PS/H2.DY, L3.PY/L1.PS/H2.YQ, L3.PY/L1.PS/H2.LT, L3.PY/L1.PS/H2.HA, L3.PY/L1.PS/H2.QL, L3.PY/L1.PS/H3.YA, L3.PY/L1.PS/H3.AE, L3.PY/L1.PS/H3.AQ, L3.PY/L1.PS/H3.TAQ, L3.PY/L1.AH/H2.QR, L3.PY/L1.AH/H2.DY, L3.PY/L1.AH/H2.YQ, L3.PY/L1.AH/H2.LT, L3.PY/L1.AH/H2.HA, L3.PY/L1.AH/H2.QL, L3.PY/L1.AH/H3.YA, L3.PY/L1.AH/H3.AE, L3.PY/L1.AH/H3.AQ, L3.PY/L1.AH/H3.TAQ, L3.PY/L1.FF/H2.QR, L3.PY/L1.FF/H2.DY, L3.PY/L1.FF/H2.YQ, L3.PY/L1.FF/H2.LT, L3.PY/L1.FF/H2.HA, L3.PY/L1.FF/H2.QL, L3.PY/L1.FF/H3.YA, L3.PY/L1.FF/H3.AE, L3.PY/L1.FF/H3.AQ, L3.PY/L1.FF/H3.TAQ, L3.PY/L1.PH/H2.QR, L3.PY/L1.PH/H2.HA, L3.PY/L1.PH/H3.AE, L3.PY/L1.PH/H3.AQ, L3.PY/L1.PH/H3.TAQ, L3.PY/L3.KY/H2.QR, L3.PY/L3.KY/H2.DY, L3.PY/L3.KY/H2.YQ L3.PY/L3.KY/H2.LT, L3.PY/L3.KY/H2.HA, L3.PY/L3.KY/H2.QL, L3.PY/L3.KY/H3.YA L3.PY/L3.KY/H3.TAQ, L3.PY/L3.KF/H2.DY, L3.PY/L3.KF/H2.YQ, L3.PY/L3.KF/H2.LT L3.PY/L3.KF/H2.QL, L3.PY/L3.KF/H3.YA, L3.PY/L3.KF/H3.AE, L3.PY/L3.KF/H3.AQ L3.PY/L3.KF/H3.TAQ, P5A2_VHVL, A02_Rd4_0.6 nM_C06, A02_Rd4_0.6 nM_C09
      • A02_Rd4_6 nM_C16, A02_Rd4_6 nM_C03, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C26
      • A02_Rd4_6 nM_C25, A02_Rd4_6 nM_C22, A02_Rd4_6 nM_C19, A02_Rd4_0.6 nM_C03
      • A02_Rd4_6 nM_C07, A02_Rd4_6 nM_C23, A02_Rd4_0.6 nM_C18, A02_Rd4_6 nM_C10
      • A02_Rd4_6 nM_C05, A02_Rd4_0.6 nM_C10, A02_Rd4_6 nM_C04, A02_Rd4_0.6 nM_C26
      • A02_Rd4_0.6 nM_C13, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C08, P5C1_VHVL, C01_Rd4_6 nM_C24, C01_Rd4_6 nM_C26, C01_Rd4_6 nM_C10, C01_Rd4_0.6 nM_C27
      • C01_Rd4_6 nM_C20, C01_Rd4_6 nM_C12, C01_Rd4_0.6 nM_C16, C01_Rd4_0.6 nM_C09
      • C01_Rd4_6 nM_C09, C01_Rd4_0.6 nM_C03, C01_Rd4_0.6 nM_C06, C01_Rd4_6 nM_C04
      • COMBO_Rd4_0.6 nM_C22, COMBO_Rd4_6 nM_C21, COMBO_Rd4_6 nM_C10, COMBO_Rd4_0.6 nM_C04, COMBO_Rd4_6 nM_C25, COMBO_Rd4_0.6 nM_C21, COMBO_Rd4_6 nM_C11, COMBO_Rd4_0.6 nM_C20, COMBO_Rd4_6 nM_C09, COMBO_Rd4_6 nM_C08, COMBO_Rd4_0.6 nM_C19, COMBO_Rd4_0.6 nM_C02, COMBO_Rd4_0.6 nM_C23, COMBO_Rd4_0.6 nM_C29, COMBO_Rd4_0.6 nM_C09, COMBO_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C30, COMBO_Rd4_0.6 nM_C14, COMBO_Rd4_6 nM_C07, COMBO_Rd4_6 nM_C02, COMBO_Rd4_0.6 nM_C05, COMBO_Rd4_0.6 nM_C17, COMBO_Rd4_6 nM_C22, COMBO_Rd4_0.6 nM_C11, COMBO_Rd4_0.6 nM_C29, P4G4, or P1A11.
  • In some embodiments, the present invention provides an antibody or an antigen binding fragment, which specifically binds to BCMA, wherein the antibody comprises a VH region comprising a sequence shown in SEQ ID NO: 112; and/or a VL region comprising a sequence shown in SEQ ID NO: 38. In some embodiments, the antibody comprises a light chain comprising the sequence EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLMYDASIRATG IPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYQSWPLTFGQGTKVEIKRTVAAPSV FIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDST YSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 357) and a heavy chain comprising the sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYPMSWVRQAPGKGLEWVSAIGGSGG SLPYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARYWPMDIWGQGTLVTV SSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPA VLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCP APELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK TKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 358).
  • In some embodiments, the present invention provides an antibody or an antigen binding fragment, which specifically bind to BCMA, wherein the antibody comprises a VH region comprising a sequence shown in SEQ ID NO: 2, 32, 42, or 78; and/or a VL region comprising a sequence shown in SEQ ID NO: 6, 16, 43, or 85.
  • In some embodiments, the invention also provides CDR portions of antibodies to BCMA antibodies based on CDR contact regions. CDR contact regions are regions of an antibody that imbue specificity to the antibody for an antigen. In general, CDR contact regions include the residue positions in the CDRs and Vernier zones which are constrained in order to maintain proper loop structure for the antibody to bind a specific antigen. See, e.g., Makabe et al., J. Biol. Chem., 283:1156-1166, 2007. Determination of CDR contact regions is well within the skill of the art.
  • The binding affinity (KD) of the BCMA antibody as described herein to BCMA (such as human BCMA (e.g., (SEQ ID NO: 353) can be about 0.002 nM to about 6500 nM. In some embodiments, the binding affinity is about any of 6500 nm, 6000 nm, 5986 nm, 5567 nm, 5500 nm, 4500 nm, 4000 nm, 3500 nm, 3000 nm, 2500 nm, 2134 nm, 2000 nm, 1500 nm, 1000 nm, 750 nm, 500 nm, 400 nm, 300 nm, 250 nm, 200 nM, 193 nM, 100 nM, 90 nM, 50 nM, 45 nM, 40 nM, 35 nM, 30 nM, 25 nM, 20 nM, 19 nm, 18 nm, 17 nm, 16 nm, 15 nM, 10 nM, 8 nM, 7.5 nM, 7 nM, 6.5 nM, 6 nM, 5.5 nM, 5 nM, 4 nM, 3 nM, 2 nM, 1 nM, 0.5 nM, 0.3 nM, 0.1 nM, 0.01 nM, or 0.002 nM. In some embodiments, the binding affinity is less than about any of 6500 nm, 6000 nm, 5500 nm, 5000 nm, 4000 nm, 3000 nm, 2000 nm, 1000 nm, 900 nm, 800 nm, 250 nM, 200 nM, 100 nM, 50 nM, 30 nM, 20 nM, 10 nM, 7.5 nM, 7 nM, 6.5 nM, 6 nM, 5 nM, 4.5 nM, 4 nM, 3.5 nM, 3 nM, 2.5 nM, 2 nM, 1.5 nM, 1 nM, or 0.5 nM.
  • In some embodiments, the invention encompasses compositions, including pharmaceutical compositions, comprising antibodies described herein or made by the methods and having the characteristics described herein. As used herein, compositions comprise one or more antibodies that bind to BCMA, and/or one or more polynucleotides comprising sequences encoding one or more these antibodies. These compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • The invention also provides methods of making any of these antibodies. The antibodies of this invention can be made by procedures known in the art. The polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis. Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available. For example, an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Pat. Nos. 5,807,715; 4,816,567; and 6,331,415.
  • The invention also encompasses fusion proteins comprising one or more fragments or regions from the antibodies of this invention. In one embodiment, a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of the variable light chain region shown in SEQ ID NOs: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38, 40, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 317, 81, 82, 84, 85, 86, 88, 89, 90, 91, 93, 94, 96, 98, 100, 102, 103, 105, 107, 108, 109, 111, 113, 115, 116, 117, 119, 121, 123, 124, 126, 128, 80, 315, 36, or 364, and/or at least 10 amino acids of the variable heavy chain region shown in SEQ ID NOs: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 112, 125, 127, 313, 314, 363, or 365. In other embodiments, a fusion polypeptide is provided that comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region. In another embodiment, the fusion polypeptide comprises a light chain variable region and/or a heavy chain variable region, as shown in any of the sequence pairs selected from among SEQ ID NOs: 1 and 2, 1 and 3, 4 and 2, 5 and 2, 6 and 2, 4 and 7, 4 and 8, 4 and 3, 9 and 8, 9 and 3, 10 and 7, 10 and 8, 10 and 3, 11 and 7, 11 and 8, 11 and 3, 12 and 3, 13 and 7, 13 and 8, 14 and 3, 15 and 2, 16 and 2, 17 and 2, 18 and 2, 19 and 2, 20 and 2, 21 and 2, 22 and 2, 23 and 2, 16 and 24, 16 and 25, 16 and 26, 16 and 27, 16 and 28, 16 and 29, 16 and 30, 16 and 31, 16 and 3, 16 and 32, 16 and 2, 18 and 24, 18 and 25, 18 and 26, 18 and 27, 18 and 28, 18 and 29, 18 and 30, 18 and 31, 18 and 3, 18 and 32, 19 and 24, 19 and 25, 19 and 26, 19 and 27, 19 and 28, 19 and 29, 19 and 30, 19 and 31, 19 and 3, 19 and 32, 20 and 24, 20 and 25, 20 and 26, 20 and 27, 20 and 28, 20 and 29, 20 and 30, 20 and 31, 20 and 3, 20 and 32, 21 and 24, 21 and 28, 21 and 31, 21 and 3, 21 and 32, 22 and 24, 22 and 25, 22 and 26, 22 and 27, 22 and 28, 22 and 29, 22 and 30, 22 and 32, 23 and 25, 23 and 26, 23 and 27, 23 and 29, 23 and 30, 23 and 31, 23 and 3, 23 and 32, 34 and 33, 36 and 35, 38 and 37, 40 and 39, 41 and 33, 43 and 42, 45 and 44, 47 and 46, 49 and 48, 51 and 50, 53 and 52, 55 and 54, 57 and 56, 59 and 58, 61 and 60, 63 and 62, 65 and 64, 67 and 66, 69 and 68, 71 and 70, 73 and 72, 75 and 74, 77 and 76, 79 and 78, 317 and 78, 79 and 78, 81 and 78, 82 and 78, 84 and 83, 85 and 78, 86 and 78, 88 and 87, 89 and 78, 90 and 78, 91 and 78, 93 and 92, 94 and 78, 96 and 95, 98 and 97, 38 and 78, 102 and 101, 103 and 78, 105 and 104, 107 and 106, 108 and 78, 109 and 78, 111 and 110, 38 and 112, 113 and 112, 115 and 114, 116 and 76, 117 and 112, 119 and 118, 121 and 120, 123 and 122, 124 and 112, 126 and 125,128 and 127, 80 and 363, or 364 and 365. In another embodiment, the fusion polypeptide comprises one or more CDR(s). In still other embodiments, the fusion polypeptide comprises CDR H3 (VH CDR3) and/or CDR L3 (VL CDR3). For purposes of this invention, a fusion protein contains one or more antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region. Exemplary heterologous sequences include, but are not limited to a “tag” such as a FLAG tag or a 6His tag. Tags are well known in the art.
  • The invention also provides isolated polynucleotides encoding the antibodies of the invention, and vectors and host cells comprising the polynucleotide.
  • In one embodiment, a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody P6E01/P6E01, P6E01/H3.AQ, L1.LGF/L3.KW/P6E01, L1.LGF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.LGF/L3.KW/H3.AL, L1.LGF/L3.KW/H3.AP, L1.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.NY/H3.AL, L1.GDF/L3.NY/H3.AP, L1.GDF/L3.NY/H3.AQ, L3.KW/P6E01,
      • L3.PY/P6E01, L3.NY/P6E01, L3.PY/L1.PS/P6E01, L3.PY/L1.AH/P6E01, L3.PY/L1.FF/P6E01, L3.PY/L1.PH/P6E01, L3.PY/L3.KY/P6E01, L3.PY/L3.KF/P6E01, L3.PY/H2.QR, L3.PY/H2.DY, L3.PY/H2.YQ, L3.PY/H2.LT, L3.PY/H2.HA, L3.PY/H2.QL, L3.PY/H3.YA, L3.PY/H3.AE, L3.PY/H3.AQ, L3.PY/H3.TAQ, L3.PY/P6E01, L3.PY/L1.PS/H2.QR, L3.PY/L1.PS/H2.DY, L3.PY/L1.PS/H2.YQ, L3.PY/L1.PS/H2.LT, L3.PY/L1.PS/H2.HA, L3.PY/L1.PS/H2.QL, L3.PY/L1.PS/H3.YA, L3.PY/L1.PS/H3.AE, L3.PY/L1.PS/H3.AQ, L3.PY/L1.PS/H3.TAQ, L3.PY/L1.AH/H2.QR, L3.PY/L1.AH/H2.DY, L3.PY/L1.AH/H2.YQ, L3.PY/L1.AH/H2.LT, L3.PY/L1.AH/H2.HA, L3.PY/L1.AH/H2.QL, L3.PY/L1.AH/H3.YA, L3.PY/L1.AH/H3.AE, L3.PY/L1.AH/H3.AQ, L3.PY/L1.AH/H3.TAQ, L3.PY/L1.FF/H2.QR, L3.PY/L1.FF/H2.DY, L3.PY/L1.FF/H2.YQ, L3.PY/L1.FF/H2.LT, L3.PY/L1.FF/H2.HA, L3.PY/L1.FF/H2.QL, L3.PY/L1.FF/H3.YA, L3.PY/L1.FF/H3.AE, L3.PY/L1.FF/H3.AQ, L3.PY/L1.FF/H3.TAQ, L3.PY/L1.PH/H2.QR, L3.PY/L1.PH/H2.HA, L3.PY/L1.PH/H3.AE, L3.PY/L1.PH/H3.AQ, L3.PY/L1.PH/H3.TAQ, L3.PY/L3.KY/H2.QR, L3.PY/L3.KY/H2.DY, L3.PY/L3.KY/H2.YQ L3.PY/L3.KY/H2.LT, L3.PY/L3.KY/H2.HA, L3.PY/L3.KY/H2.QL, L3.PY/L3.KY/H3.YA L3.PY/L3.KY/H3.TAQ, L3.PY/L3.KF/H2.DY, L3.PY/L3.KF/H2.YQ, L3.PY/L3.KF/H2.LT L3.PY/L3.KF/H2.QL, L3.PY/L3.KF/H3.YA, L3.PY/L3.KF/H3.AE, L3.PY/L3.KF/H3.AQ L3.PY/L3.KF/H3.TAQ, P5A2_VHVL, A02_Rd4_0.6 nM_C06, A02_Rd4_0.6 nM_C09
      • A02_Rd4_6 nM_C16, A02_Rd4_6 nM_C03, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C26
      • A02_Rd4_6 nM_C25, A02_Rd4_6 nM_C22, A02_Rd4_6 nM_C19, A02_Rd4_0.6 nM_C03
      • A02_Rd4_6 nM_C07, A02_Rd4_6 nM_C23, A02_Rd4_0.6 nM_C18, A02_Rd4_6 nM_C10
      • A02_Rd4_6 nM_C05, A02_Rd4_0.6 nM_C10, A02_Rd4_6 nM_C04, A02_Rd4_0.6 nM_C26
      • A02_Rd4_0.6 nM_C13, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C08, P5C1_VHVL, C01_Rd4_6 nM_C24, C01_Rd4_6 nM_C26, C01_Rd4_6 nM_C10, C01_Rd4_0.6 nM_C27
      • C01_Rd4_6 nM_C20, C01_Rd4_6 nM_C12, C01_Rd4_0.6 nM_C16, C01_Rd4_0.6 nM_C09
      • C01_Rd4_6 nM_C09, C01_Rd4_0.6 nM_C03, C01_Rd4_0.6 nM_C06, C01_Rd4_6 nM_C04
      • COMBO_Rd4_0.6 nM_C22, COMBO_Rd4_6 nM_C21, COMBO_Rd4_6 nM_C10, COMBO_Rd4_0.6 nM_C04, COMBO_Rd4_6 nM_C25, COMBO_Rd4_0.6 nM_C21, COMBO_Rd4_6 nM_C11, COMBO_Rd4_0.6 nM_C20, COMBO_Rd4_6 nM_C09, COMBO_Rd4_6 nM_C08, COMBO_Rd4_0.6 nM_C19, COMBO_Rd4_0.6 nM_C02, COMBO_Rd4_0.6 nM_C23, COMBO_Rd4_0.6 nM_C29, COMBO_Rd4_0.6 nM_C09, COMBO_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C30, COMBO_Rd4_0.6 nM_C14, COMBO_Rd4_6 nM_C07, COMBO_Rd4_6 nM_C02, COMBO_Rd4_0.6 nM_C05, COMBO_Rd4_0.6 nM_C17, COMBO_Rd4_6 nM_C22, COMBO_Rd4_0.6 nM_C11, COMBO_Rd4_0.6 nM_C29, P4G4, or P1A11. The sequence encoding the antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use. Vectors (including expression vectors) and host cells are further described herein.
  • The invention also encompasses scFv of antibodies of this invention. Single chain variable region fragments are made by linking light and/or heavy chain variable regions by using a short linking peptide (Bird et al., Science 242:423-426, 1988). An example of a linking peptide is (GGGGS)3 (SEQ ID NO: 498), which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region. Linkers of other sequences have been designed and used (Bird et al., 1988, supra). Linkers should be short, flexible polypeptides and preferably comprised of less than about 20 amino acid residues. Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports. The single chain variants can be produced either recombinantly or synthetically. For synthetic production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli. Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides. The resultant scFv can be isolated using standard protein purification techniques known in the art.
  • Other forms of single chain antibodies, such as diabodies or minibodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which heavy chain variable (VH) and light chain variable (VL) domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al., Proc. Natl. Acad Sci. USA 90:6444-6448, 1993; Poljak, R. J., et al., Structure 2:1121-1123, 1994). Minibody includes the VL and VH domains of a native antibody fused to the hinge region and CH3 domain of the immunoglobulin molecule. See, e.g., U.S. Pat. No. 5,837,821.
  • In another aspect, the invention provides compositions (such as a pharmaceutical compositions) comprising any of the polynucleotides of the invention. In some embodiments, the composition comprises an expression vector comprising a polynucleotide encoding any of the antibodies described herein. In still other embodiments, the composition comprises either or both of the polynucleotides shown in SEQ ID NO: 486 and SEQ ID NO: 485 below:
  • COMBO_Rd4_0.6nM_C29 heavy chain variable region
    (SEQ ID NO: 486)
    GAAGTCCAACTCCTCGAATCCGGTGGCGGCCTTGTCCAGCCTGGAGGTT
    CCTTGCGCCTGTCATGTGCCGCCAGCGGATTCACCTTCTCGTCCTACCC
    GATGTCGTGGGTCCGCCAGGCTCCGGGAAAGGGCCTGGAATGGGTGTCA
    GCCATCGGAGGATCGGGGGGCTCCCTGCCCTACGCCGATATCGTGAAGG
    GAAGGTTCACCATTAGCCGGGACAACTCCAAGAACACTCTGTACCTCCA
    AATGAACAGCCTGAGAGCGGAGGACACCGCAGTGTACTATTGCGCCCGG
    TACTGGCCAATGGACATCTGGGGCCAGGGGACTCTGGTCACCGTCTCCT
    CA
    COMBO_Rd4_0.6nM_C29 light chain variable region
    (SEQ ID NO: 485)
    GAGATCGTGCTGACTCAGTCCCCTGGAACCCTGTCCCTGTCACCTGGCG
    AAAGAGCTACCTTGTCCTGTCGCGCATCACAATCCGTGTCGTCGAGCTA
    TCTCGCGTGGTACCAGCAGAAGCCCGGACAGGCCCCAAGGCTGCTTATG
    TACGACGCCTCCATCCGGGCCACTGGTATCCCCGACCGCTTCTCGGGCT
    CCGGAAGCGGCACCGACTTCACCCTGACTATTTCCCGGCTCGAACCGGA
    GGATTTCGCCGTGTACTACTGCCAACAGTACCAGAGCTGGCCGCTGACG
    TTTGGGCAGGGGACCAAGGTCGAAATCAAA
  • In other embodiments, the composition comprises either or both of the polynucleotides shown in SEQ ID NO: 488 and SEQ ID NO: 487 below:
  • L3.PY/H3TAQ heavy chain variable region
    (SEQ ID NO: 488)
    GAAGTGCAGCTGCTGGAATCTGGCGGAGGACTGGTGCAGCCTGGCGGCT
    CTCTGAGACTGTCTTGTGCCGCCAGCGGCTTCACCTTCGGCAGCTACGC
    TATGACCTGGGTGCGCCAGGCCCCTGGCAAAGGACTGGAATGGGTGTCC
    GCCATCTCTGGCAGCGGCGGCAATACCTTCTACGCCGAGAGCGTGAAGG
    GCCGGTTCACCATCAGCCGGGACAACAGCAAGAACACCCTGTACCTGCA
    GATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGTACACGG
    GTGTCCCCTATCGCCGCGCAGATGGATTATTGGGGCCAGGGCACTCTGG
    TCACCGTCTCCTCA
    L3.PY/H3TAQ heavy chain variable region
    (SEQ ID NO: 487)
    GAGATCGTGCTGACACAGAGCCCTGGCACCCTGAGCCTGTCTCCAGGCG
    AAAGAGCCACCCTGTCCTGCAGAGCCAGCCAGAGCGTGTCCAGCAGCTA
    CCTGGCCTGGTATCAGCAGAAGCCCGGCCAGGCTCCCCGGCTGCTGATC
    TATGGCGCCTCTTCTAGAGCCACCGGCATCCCCGATAGATTCAGCGGCT
    CTGGCAGCGGCACCGACTTCACCCTGACCATCAGCAGACTGGAACCCGA
    GGACTTCGCCGTGTACTACTGCCAGCACTACCCTTATCCCCCCAGCTTC
    ACATTTGGCCAGGGCACCAAGGTGGAGATCAAA
  • In still other embodiments, the composition comprises either or both of the polynucleotides shown in SEQ ID NO: 490 and SEQ ID NO: 489 below:
  • A02_Rd4_0.6nM_C01 heavy chain variable region
    (SEQ ID NO: 490)
    GAAGTTCAATTATTGGAATCTGGTGGAGGACTGGTGCAGCCTGGCGGCT
    CTCTGAGACTGTCTTGTGCCGCCAGCGGCTTCACCTTCAGCAGCTACGC
    CATGAACTGGGTGCGCCAGGCCCCTGGTAAAGGTTTGGAATGGGTTTCT
    GCTATTACTGCGTCTGGTGGTTCTACTTACTATGCCGATGTGGTTAAGG
    GTAGATTCACCATTTCTAGAGACAACTCTAAGAACACCTTGTACTTGCA
    AATGAACTCCTTGAGAGCTGAAGATACTGCTGTTTATTACTGTGCTAGA
    TACTGGCCAATGTCGTTGTGGGGTCAAGGTACTCTGGTCACCGTCTCCT
    CA
    A02_Rd4_0.6nM_C01 light chain variable region
    (SEQ ID NO: 489)
    GAGATCGTGCTGACACAGAGCCCTGGCACCCTGAGCCTGTCTCCTGGTG
    AAAGAGCTACTTTGTCTTGTAGAGCTTCTCAATCCGTTTCCGCGTATTA
    TTTGGCTTGGTATCAACAAAAACCAGGTCAAGCTCCAAGATTATTGATG
    TACGATGCTTCTATTAGAGCCACCGGTATTCCAGATAGATTTTCTGGTT
    CTGGTTCCGGTACTGATTTCACTTTGACTATCTCTAGATTGGAACCAGA
    AGATTTCGCTGTTTACTACTGTCAACAATATGAGCGTTGGCCATTGACT
    TTTGGTCAAGGTACAAAGGTTGAAATCAAACGTGAG
  • In other embodiments, the composition comprises either or both of the polynucleotides shown in SEQ ID NO: 492 and SEQ ID NO: 491 below:
  • A02_Rd4_0.6nM_C16 heavy chain variable region
    (SEQ ID NO: 492)
    GAAGTTCAATTATTGGAATCTGGTGGAGGACTGGTGCAGCCTGGCGGCT
    CTCTGAGACTGTCTTGTGCCGCCAGCGGCTTCACCTTCAGCAGCTACGC
    CATGAACTGGGTGCGCCAGGCCCCTGGTAAAGGTTTGGAATGGGTTTCT
    GCTATTTCTGATTTTGGTGGTTCTACTTACTATGCCGATATCGTTAAGG
    GTAGATTCACCATTTCTAGAGACAACTCTAAGAACACCTTGTACTTGCA
    AATGAACTCCTTGAGAGCTGAAGATACTGCTGTTTATTACTGTGCTAGA
    TACTGGCCAATGGATATTTGGGGTCAAGGTACTCTGGTCACCGTCTCCT
    CA
    A02_Rd4_0.6nM_C16 light chain variable region
    (SEQ ID NO: 491)
    GAGATCGTGCTGACACAGAGCCCTGGCACCCTGAGCCTGTCTCCTGGTG
    AAAGAGCTACTTTGTCTTGTAGAGCTTCTCAATCCGTTTCCGATCTGTA
    TTTGGCTTGGTATCAACAAAAACCAGGTCAAGCTCCAAGATTATTGATG
    TACGATGCTTCTATTAGAGCCACCGGTATTCCAGATAGATTTTCTGGTT
    CTGGTTCCGGTACTGATTTCACTTTGACTATCTCTAGATTGGAACCAGA
    AGATTTCGCTGTTTACTACTGTCAACAATATCAGACTTGGCCATTGACT
    TTTGGTCAAGGTACAAAGGTTGAAATCAAACGTGAG.
  • Expression vectors, and administration of polynucleotide compositions are further described herein.
  • In another aspect, the invention provides a method of making any of the polynucleotides described herein.
  • Polynucleotides complementary to any such sequences are also encompassed by the present invention. Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules. RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence. Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein. Variants preferably exhibit at least about 70% identity, more preferably, at least about 80% identity, yet more preferably, at least about 90% identity, and most preferably, at least about 95% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A “comparison window” as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O., 1978, A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D. G. and Sharp, P. M., 1989, CABIOS 5:151-153; Myers, E. W. and Muller W., 1988, CABIOS 4:11-17; Robinson, E. D., 1971, Comb. Theor. 11:105; Santou, N., Nes, M., 1987, Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R., 1973, Numerical Taxonomy the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, CA; Wilbur, W. J. and Lipman, D. J., 1983, Proc. Natl. Acad. Sci. USA 80:726-730.
  • Preferably, the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof. Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence).
  • Suitable “moderately stringent conditions” include prewashing in a solution of 5×SSC. 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-65° C., 5×SSC, overnight; followed by washing twice at 65° C. for 20 minutes with each of 2×, 0.5× and 0.2×SSC containing 0.1% SDS.
  • As used herein, “highly stringent conditions” or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42° C., with washes at 42° C. in 0.2×SSC (sodium chloride/sodium citrate) and 50% formamide at 55° C., followed by a high-stringency wash consisting of 0.1×SSC containing EDTA at 55° C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
  • It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • The polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
  • For preparing polynucleotides using recombinant methods, a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein. Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome. The polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al., 1989.
  • Alternatively, PCR allows reproduction of DNA sequences. PCR technology is well known in the art and is described in U.S. Pat. Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston, 1994.
  • RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., 1989, supra, for example.
  • Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector. Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, pMB9, CoIE1, pCR1, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462. Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • The vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus). The choice of introducing vectors or polynucleotides will often depend on features of the host cell.
  • The invention also provides host cells comprising any of the polynucleotides described herein. Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest. Non-limiting examples of mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462. Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe; or K. lactis). Preferably, the host cells express the cDNAs at a level of about 5 fold higher, more preferably, 10 fold higher, even more preferably, 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells. Screening the host cells for a specific binding to BCMA or an BCMA domain (e.g., domains 1-4) is effected by an immunoassay or FACS. A cell overexpressing the antibody or protein of interest can be identified.
  • Representative materials of the present invention were deposited in the American Type Culture Collection (ATCC) on Apr. 15, 2015. Vector having ATCC Accession No.
  • PTA-122094 is a polynucleotide encoding a humanized BCMA antibody heavy chain variable region, and vector having ATCC Accession No. PTA-122093 is a polynucleotide encoding a humanized BCMA antibody light chain variable region. The deposits were made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and Regulations thereunder (Budapest Treaty). This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit. The deposit will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Pfizer, Inc. and ATCC, which assures permanent and unrestricted availability of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U.S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 U.S.C. Section 122 and the Commissioner's rules pursuant thereto (including 37 C.F.R. Section 1.14 with particular reference to 886 OG 638).
  • The assignee of the present application has agreed that if a culture of the materials on deposit should die or be lost or destroyed when cultivated under suitable conditions, the materials will be promptly replaced on notification with another of the same. Availability of the deposited material is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws.
  • BCMA Antibody Conjugates
  • The present invention also provides a conjugate (or immunoconjugate) of the BCMA antibody as described herein, or of the antigen binding fragment thereof, wherein the antibody or the antigen binding fragment is conjugated to an agent (e.g., a cytotoxic agent) for targeted immunotherapy (e.g., antibody-drug conjugates) either directly or indirectly via a linker. For example, a cytotoxic agent can be linked or conjugated to the BCMA antibody or the antigen binding fragment thereof as described herein for targeted local delivery of the cytotoxic agent moiety to tumors (e.g., BCMA expressing tumor).
  • Methods for conjugating cytotoxic agent or other therapeutic agents to antibodies have been described in various publications. For example, chemical modification can be made in the antibodies either through lysine side chain amines or through cysteine sulfhydryl groups activated by reducing interchain disulfide bonds for the conjugation reaction to occur. See, e.g., Tanaka et al., FEBS Letters 579:2092-2096, 2005, and Gentle et al., Bioconjugate Chem. 15:658-663, 2004. Reactive cysteine residues engineered at specific sites of antibodies for specific drug conjugation with defined stoichiometry have also been described. See, e.g., Junutula et al., Nature Biotechnology, 26:925-932, 2008. Conjugation using an acyl donor glutamine-containing tag or an endogenous glutamine made reactive (i.e., the ability to form a covalent bond as an acyl donor) by polypeptide engineering in the presence of transglutaminase and an amine (e.g., a cytotoxic agent comprising or attached to a reactive amine) is also described in international applications WO2012/059882 and WO2015015448.
  • In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody (e.g., a carboxyl terminus, an amino terminus, or at another site in the BCMA antibody). In some embodiments, the tag comprises an amino acid glutamine (Q) or an amino acid sequence LQG, LLQGG (SEQ ID NO:318), LLQG (SEQ ID NO:454), LSLSQG (SEQ ID NO: 455), GGGLLQGG (SEQ ID NO: 456), GLLQG (SEQ ID NO: 457), LLQ, GSPLAQSHGG (SEQ ID NO: 458), GLLQGGG (SEQ ID NO: 459), GLLQGG (SEQ ID NO: 460), GLLQ (SEQ ID NO: 461), LLQLLQGA (SEQ ID NO: 462), LLQGA (SEQ ID NO: 463), LLQYQGA (SEQ ID NO: 464), LLQGSG (SEQ ID NO: 465), LLQYQG (SEQ ID NO: 466), LLQLLQG (SEQ ID NO: 467), SLLQG (SEQ ID NO: 468), LLQLQ (SEQ ID NO: 469), LLQLLQ (SEQ ID NO: 470), LLQGR (SEQ ID NO: 471), LLQGPP (SEQ ID NO: 472), LLQGPA (SEQ ID NO: 473), GGLLQGPP (SEQ ID NO: 474), GGLLQGA (SEQ ID NO: 475), LLQGPGK (SEQ ID NO: 476), LLQGPG (SEQ ID NO: 477), LLQGP (SEQ ID NO: 478), LLQP (SEQ ID NO: 479), LLQPGK (SEQ ID NO: 480), LLQAPGK (SEQ ID NO: 481), LLQGAPG (SEQ ID NO: 482), LLQGAP (SEQ ID NO: 483), and LLQLQG (SEQ ID NO: 484).
  • In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody, wherein the tag comprises an amino acid sequence GGLLQGPP (SEQ ID NO: 474) or GGLLQGA (SEQ ID NO: 475) engineered at the light chain carboxyl terminus of the BCMA antibody. In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody, wherein the tag comprises an amino acid sequence LLQG (SEQ ID NO: 454) engineered after residue T135 in the heavy chain of the BCMA antibody. In other embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag engineered at a specific site of the antibody, wherein the tag comprises an amino acid sequence LLQGA (SEQ ID NO: 463) or LLQGPP (SEQ ID NO: 472) engineered at the heavy chain carboxyl terminus of the BCMA antibody and wherein the lysine residue at the heavy chain carboxyl terminus is deleted. In some embodiments, the BCMA antibody or the conjugate as described herein comprises an amino acid substitution at position 297 of the BCMA antibody (EU numbering scheme). For example, the amino acid asparagine (N) can be substituted with glutamine (Q) or alanine (A) at position 297 of the BCMA antibody.
  • Also provided is an isolated antibody comprising an acyl donor glutamine-containing tag and an amino acid modification at position 222, 340, or 370 of the antibody (EU numbering scheme) wherein the modification is an amino acid deletion, insertion, substitution, mutation, or any combination thereof. Accordingly, in some embodiments, provided is the BCMA antibody or the conjugate as described herein comprising the acyl donor glutamine-containing tag (e.g., Q, LQG, LLQGG (SEQ ID NO:318), LLQG (SEQ ID NO:454), LSLSQG (SEQ ID NO: 455), GGGLLQGG (SEQ ID NO: 456), GLLQG (SEQ ID NO: 457), LLQ, GSPLAQSHGG (SEQ ID NO: 458), GLLQGGG (SEQ ID NO: 459), GLLQGG (SEQ ID NO: 460), GLLQ (SEQ ID NO: 461), LLQLLQGA (SEQ ID NO: 462), LLQGA (SEQ ID NO: 463), LLQYQGA (SEQ ID NO: 464), LLQGSG (SEQ ID NO: 465), LLQYQG (SEQ ID NO: 466), LLQLLQG (SEQ ID NO: 467), SLLQG (SEQ ID NO: 468), LLQLQ (SEQ ID NO: 469), LLQLLQ (SEQ ID NO: 470), LLQGR (SEQ ID NO: 471), LLQGPP (SEQ ID NO: 472), LLQGPA (SEQ ID NO: 473), GGLLQGPP (SEQ ID NO: 474), GGLLQGA (SEQ ID NO: 475), LLQGPGK (SEQ ID NO: 476), LLQGPG (SEQ ID NO: 477), LLQGP (SEQ ID NO: 478), LLQP (SEQ ID NO: 479), LLQPGK (SEQ ID NO: 480), LLQAPGK (SEQ ID NO: 481), LLQGAPG (SEQ ID NO: 482), LLQGAP (SEQ ID NO: 483), and LLQLQG (SEQ ID NO: 484)) conjugated at a specific site (e.g., at a carboxyl terminus of the heavy or light chain, residue T135 in the antibody heavy chain, or at another site) of the BCMA antibody and an amino acid modification at position 222, 340, or 370 of the antibody (EU numbering scheme). In some embodiments, the amino acid modification is a substitution from lysine to arginine (e.g., K222R, K340R, or K370R).
  • In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence GGLLQGPP (SEQ ID NO: 474) engineered at the C-terminus of the BCMA antibody light chain and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme). In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence GGLLQGA (SEQ ID NO: 475) engineered at the C-terminus of the BCMA antibody light chain and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme). In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence LLQGA (SEQ ID NO: 463) engineered at the C-terminus of the BCMA antibody heavy chain and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme), wherein the lysine residue at the heavy chain carboxyl terminus is deleted. In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence LLQG (SEQ ID NO: 454) engineered after residue T135 in the heavy chain of the BCMA antibody and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising a glutamine engineered at position 297 or an amino acid substitution at position 297 from asparagine (N) to another amino acid in the BCMA antibody and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme). For example, in some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence GGLLQGPP (SEQ ID NO: 474) engineered at the C-terminus of the BCMA antibody light chain, an amino acid substitution at position 297 of the BCMA antibody from asparagine (N) to glutamine (Q), and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme). In some embodiments, the BCMA antibody or the conjugate as described herein comprises an acyl donor glutamine-containing tag comprising the sequence LLQG (SEQ ID NO: 454) engineered after residue T135 in the heavy chain of the BCMA antibody, an amino acid substitution at position 297 of the BCMA antibody from asparagine (N) to alanine (A), and an amino acid substitution from lysine to arginine at position 222 of the antibody (EU numbering scheme).
  • The agents that can be conjugated to the BCMA antibodies or the antigen binding fragments of the present invention include, but are not limited to, cytotoxic agents, immunomodulating agents, imaging agents, therapeutic proteins, biopolymers, or oligonucleotides.
  • Examples of a cytotoxic agent include, but are not limited to, anthracycline, an auristatin, a dolastatin, a combretastatin, a duocarmycin, a pyrrolobenzodiazepine dimer, an indolino-benzodiazepine dimer, an enediyne, a geldanamycin, a maytansine, a puromycin, a taxane, a vinca alkaloid, a camptothecin, a tubulysin, a hemiasterlin, a spliceostatin, a pladienolide, and stereoisomers, isosteres, analogs, or derivatives thereof.
  • The anthracyclines are derived from bacteria Strepomyces and have been used to treat a wide range of cancers, such as leukemias, lymphomas, breast, uterine, ovarian, and lung cancers. Exemplary anthracyclines include, but are not limited to, daunorubicin, doxorubicin (i.e., adriamycin), epirubicin, idarubicin, valrubicin, and mitoxantrone.
  • Dolastatins and their peptidic analogs and derivatives, auristatins, are highly potent antimitotic agents that have been shown to have anticancer and antifungal activity. See, e.g., U.S. Pat. No. 5,663,149 and Pettit et al., Antimicrob. Agents Chemother. 42:2961-2965, 1998. Exemplary dolastatins and auristatins include, but are not limited to, dolastatin 10, auristatin E, auristatin EB (AEB), auristatin EFP (AEFP), MMAD (Monomethyl Auristatin D or monomethyl dolastatin 10), MMAF (Monomethyl Auristatin F or N-methylvaline-valine-dolaisoleuine-dolaproine-phenylalanine), MMAE (Monomethyl Auristatin E or N-methylvaline-valine-dolaisoleuine-dolaproine-norephedrine), 5-benzoylvaleric acid-AE ester (AEVB), and other novel auristatins (such as the ones described in U.S. Publication No. 2013/0129753). In some embodiments, the auristatin is 0101 (2-methylalanyl-N-[(3R,4S,5S)-3-methoxy-1-{(2S)-2-[(1R,2R)-1-methoxy-2-methyl-3-oxo-3-{[(1 S)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino}propyl]pyrrolidin-1-yl}-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide) having the following structure:
  • Figure US20240018252A1-20240118-C00001
  • In some embodiments, the auristatin is 3377 (N,2-dimethylalanyl-N-{(1 S,2R)-4-{(2S)-2-[(1R,2R)-3-{[(1S)-1-carboxyl-2-phenylethyl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-2-methoxy-1-[(1 S)-1-methylpropyl]-4-oxobutyl}-N-methyl-L-valinamide) having the following structure:
  • Figure US20240018252A1-20240118-C00002
  • In some embodiments, the auristatin is 0131-OMe (N,2-dimethylalanyl-N-[(3R,4S,5S)-3-methoxy-1-{(2S)-2-[(1R,2R)-1-methoxy-3-{[(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino}-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-5-methyl-1-oxoheptan-4-yl]-N-methylL-valinamide) having the following structure:
  • Figure US20240018252A1-20240118-C00003
  • In other embodiments, the auristatin is 0131 (2-methyl-L-prolyl-N-[(3R,4S,5S)-1-{(2S)-2-[(1R,2R)-3-{[(1S)-1-carboxy-2-phenylethyl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide) having the following structure:
  • Figure US20240018252A1-20240118-C00004
  • In other embodiments, the auristatin is 0121 (2-methyl-L-proly-N-[(3R,4S,5S)-1-{(2S)-2-[(1R,2R)-3-{[(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide) having the following structure:
  • Figure US20240018252A1-20240118-C00005
  • Camptothecin is a cytotoxic quinoline alkaloid which inhibits the enzyme topoisomerase I. Examples of camptothecin and its derivatives include, but are not limited to, topotecan and irinotecan, and their metabolites, such as SN-38.
  • Combretastatins are natural phenols with vascular disruption properties in tumors. Exemplary combretastatins and their derivatives include, but are not limited to, combretastatin A-4 (CA-4) and ombrabulin.
  • Duocarmycin and CC-1065 are DNA alkylating agents with cytotoxic potency. See Boger and Johnson, PNAS 92:3642-3649 (1995). Exemplary duocarmycin and CC-1065 include, but are not limited to, (+)-duocarmycin A and (+)-duocarmycin SA, (+)-CC-1065, and the compounds as disclosed in the international application PCT/IB2015/050280 including, but not limited to, N˜2˜acetyl-L-lysyl-L-valyl-N˜5˜carbamoyl-N-[4-({[(2-{[({(1S)-1-(chloromethyl)-3-[(5-{[(1S)-1-(chloromethyl)-5-(phosphonooxy)-1,2-dihydro-3H-benzo[e]indol-3-yl]carbonyl}thiophen-2-yl)carbonyl]-2,3-dihydro-1H-benzo[e]indol-5-yl}oxy)carbonyl](methyl)amino}ethyl)(methyl)carbamoyl]oxy}methyl)phenyl]-L-ornithinamide having the structure:
  • Figure US20240018252A1-20240118-C00006
  • N˜2˜acetyl-L-lysyl-L-valyl-N˜5˜carbamoyl-N-[4-({[(2-{[({(8S)-8-(chloromethyl)-6-[(3-{[(1 S)-1-(chloromethyl)-8-methyl-5-(phosphonooxy)-1,6-dihydropyrrolo[3,2-e]indol-3(2H)-yl]carbonyl}bicyclo[1.1.1]pent-1-yl)carbonyl]-1-methyl-3,6,7,8-tetrahydropyrrolo[3,2-e]indol-4-yl}oxy)carbonyl](methyl)amino}ethyl)(methyl)carbamoyl]oxy}methyl)phenyl]-L-ornithinamide having the structure:
  • Figure US20240018252A1-20240118-C00007
  • N˜2˜acetyl-L-lysyl-L-valyl-N˜5˜carbamoyl-N-[4-({[(2-{[({(8S)-8-(chloromethyl)-6-[(4-{[(1 S)-1-(chloromethyl)-8-methyl-5-(phosphonooxy)-1,6-dihydropyrrolo[3,2-e]indol-3(2H)-yl]carbonyl}pentacyclo[4.2.0.0-2,5˜.0˜3,8-.0˜4,7˜]oct-1-yl)carbonyl]-1-methyl-3,6,7,8-tetrahydropyrrolo[3,2-e]indol-4-yl}oxy)carbonyl](methyl)amino}ethyl)(methyl)carbamoyl]oxy}methyl)phenyl]-L-ornithinamide having the structure:
  • Figure US20240018252A1-20240118-C00008
  • Enediynes are a class of anti-tumor bacterial products characterized by either nine- and ten-membered rings or the presence of a cyclic system of conjugated triple-double-triple bonds. Exemplary enediynes include, but are not limited to, calicheamicin, esperamicin, uncialamicin, dynemicin, and their derivatives.
  • Geldanamycins are benzoquinone ansamycin antibiotic that bind to Hsp90 (Heat Shock Protein 90) and have been used antitumor drugs. Exemplary geldanamycins include, but are not limited to, 17-AAG (17-N-Allylamino-17-Demethoxygeldanamycin) and 17-DMAG (17-Dimethylaminoethylamino-17-demethoxygeldanamycin).
  • Hemiasterlin and its analogues (e.g., HTI-286) bind to the tubulin, disrupt normal microtubule dynamics, and, at stoichiometric amounts, depolymerize microtubules.
  • Maytansines or their derivatives maytansinoids inhibit cell proliferation by inhibiting the microtubules formation during mitosis through inhibition of polymerization of tubulin. See Remillard et al., Science 189:1002-1005, 1975. Exemplary maytansines and maytansinoids include, but are not limited to, mertansine (DM1) and its derivatives as well as ansamitocin.
  • Pyrrolobenzodiazepine dimers (PBDs) and indolino-benzodiazepine dimers (IGNs) are anti-tumor agents that contain one or more immine functional groups, or their equivalents, that bind to duplex DNA. PBD and IGN molecules are based on the natural product athramycin, and interact with DNA in a sequence-selective manner, with a preference for purine-guanine-purine sequences. Exemplary PBDs and their analogs include, but are not limited to, SJG-136.
  • Spliceostatins and pladienolides are anti-tumor compounds which inhibit splicing and interacts with spliceosome, SF3b. Examples of spliceostatins include, but are not limited to, spliceostatin A, FR901464, and (2S,3Z)-5-{[(2R,3R,5S,6S)-6-{(2E,4E)-5-[(3R,4R,5R,7S)-7-(2-hydrazinyl-2-oxoethyl)-4-hydroxy-1,6-dioxaspiro[2.5]oct-5-yl]-3-methylpenta-2,4-dien-1-yl}-2,5-dimethyltetrahydro-2H-pyran-3-yl]amino}-5-oxopent-3-en-2-yl acetate having the structure of
  • Figure US20240018252A1-20240118-C00009
  • Examples of pladienolides include, but are not limited to, Pladienolide B, Pladienolide D, or E7107.
  • Taxanes are diterpenes that act as anti-tubulin agents or mitotic inhibitors. Exemplary taxanes include, but are not limited to, paclitaxel (e.g., TAXOL®) and docetaxel (TAXOTERE®).
  • Tubulysins are natural products isolated from a strain of myxobacteria that has been shown to depolymerize microtubules and induce mitotic arrest. Exemplary tubulysins include, but are not limited to, tubulysin A, tubulysin B, and tubulysin D.
  • Vinca alkyloids are also anti-tubulin agents. Exemplary vinca alkyloids include, but are not limited to, vincristine, vinblastine, vindesine, and vinorelbine.
  • Accordingly, in some embodiments, the cytotoxic agent is selected from the group consisting of MMAD (Monomethyl Auristatin D), 0101 (2-methylalanyl-N-[(3R,4S,5S)-3-methoxy-1-{(2S)-2-[(1R,2R)-1-methoxy-2-methyl-3-oxo-3-{[(1 S)-2-phenyl-1-(1,3-thiazol-2-yl)ethyl]amino}propyl]pyrrolidin-1-yl}-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), 3377 (N,2-dimethylalanyl-N-{(1 S,2R)-4-{(2S)-2-[(1R,2R)-3-{[(1 S)-1-carboxyl-2-phenylethyl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrroidin-1-yl}-2-methoxy-1-[(1 S)-1-methylpropyl]-4-oxobutyl}-N-methyl-L-valinamide), 0131 (2-methyl-L-proly-N-[(3R,4S,5S)-1-{(2S)-2-[(1R,2R)-3-{[(1 S)-1-carboxy-2-phenylethyl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), 0131-OMe (N,2-dimethylalanyl-N-[(3R,4S,5S)-3-methoxy-1-{(2S)-2-[(1R,2R)-1-methoxy-3-{[(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino}-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-5-methyl-1-oxoheptan-4-yl]-N-methylL-valinamide), 0121(2-methyl-L-proly-N-[(3R,4S,5S)-1-{(2S)-2-[(1R,2R)-3-{[(2S)-1-methoxy-1-oxo-3-phenylpropan-2-yl]amino}-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl}-3-methoxy-5-methyl-1-oxoheptan-4-yl]-N-methyl-L-valinamide), and (2S,3Z)-5-{[(2R,3R,5S,6S)-6-{(2E,4E)-5-[(3R,4R,5R,7S)-7-(2-hydrazinyl-2-oxoethyl)-4-hydroxy-1,6-dioxaspiro[2.5]oct-5-yl]-3-methylpenta-2,4-dien-1-yl}-2,5-dimethyltetrahydro-2H-pyran-3-yl]amino}-5-oxopent-3-en-2-yl acetate.
  • In some embodiments, the agent is an immunomodulating agent. Examples of an immunomodulating agent include, but are not limited to, gancyclovier, etanercept, tacrolimus, sirolimus, voclosporin, cyclosporine, rapamycin, cyclophosphamide, azathioprine, mycophenolgate mofetil, methotrextrate, glucocorticoid and its analogs, cytokines, stem cell growth factors, lymphotoxins, tumor necrosis factor (TNF), hematopoietic factors, interleukins (e.g., interleukin-1 (IL-1), IL-2, IL-3, IL-6, IL-10, IL-12, IL-18, and IL-21), colony stimulating factors (e.g., granulocyte-colony stimulating factor (G-CSF) and granulocyte macrophage-colony stimulating factor (GM-CSF)), interferons (e.g., interferons-α, -β and -γ), the stem cell growth factor designated “S 1 factor,” erythropoietin and thrombopoietin, or a combination thereof.
  • In some embodiments, the agent moiety is an imaging agent (e.g., a fluorophore or a chelator), such as fluorescein, rhodamine, lanthanide phosphors, and their derivatives thereof, or a radioisotope bound to a chelator. Examples of fluorophores include, but are not limited to, fluorescein isothiocyanate (FITC) (e.g., 5-FITC), fluorescein amidite (FAM) (e.g., 5-FAM), eosin, carboxyfluorescein, erythrosine, Alexa Fluor® (e.g., Alexa 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 647, 660, 680, 700, or 750), carboxytetramethylrhodamine (TAMRA) (e.g., 5-TAMRA), tetramethylrhodamine (TMR), and sulforhodamine (SR) (e.g., SR101). Examples of chelators include, but are not limited to, 1,4,7,10-tetraazacyclododecane-N,N′,N″,N′″-tetraacetic acid (DOTA), 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (deferoxamine), diethylenetriaminepentaacetic acid (DTPA), and 1,2-bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid) (BAPTA).
  • Examples of fluorophores include, but are not limited to, fluorescein isothiocyanate (FITC) (e.g., 5-FITC), fluorescein amidite (FAM) (e.g., 5-FAM), eosin, carboxyfluorescein, erythrosine, Alexa Fluor® (e.g., Alexa 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 647, 660, 680, 700, or 750), carboxytetramethylrhodamine (TAMRA) (e.g., 5-TAMRA), tetramethylrhodamine (TMR), and sulforhodamine (SR) (e.g., SR101).
  • In some embodiments, therapeutic or diagnostic radioisotopes or other labels (e.g., PET or SPECT labels) can be incorporated in the agent for conjugation to the BCMA antibodies or the antigen binding fragments as described herein. Examples of a radioisotope or other labels include, but are not limited to, 3H, 11C, 13N, 14C, 15N, 15O, 35S, 18F 32P, 33P, 47Sc, 51Cr, 57Co, 58Co, 59Fe, 62Cu, 64Cu, 67Cu, 67Ga, 68Ga, 75Se, 76Br, 77Br, 86Y 89Zr, 90Y, 94Tc, 95Ru, 97Ru, 99Tc, 103Ru, 105Rh, 105Ru, 107Hg, 109Pd, 111Ag, 111In, 113In, 121Te, 122Te, 123I, 124I, 125I, 125Te, 126I, 131I, 131In, 133I, 142Pr, 143Pr, 153Pb, 153Sm, 161Tb, 165Tm, 166Dy, 166H, 167Tm, 168Tm, 169Yb, 177Lu, 186Re, 188Re, 189Re, 197Pt, 198Au, 199Au, 201Tl, 203Hg, 211At, 212Bi, 212Pb, 213Bi, 223Ra, 224Ac, or 225Ac.
  • In some embodiments, the agent is a therapeutic protein including, but is not limited to, a toxin, a hormone, an enzyme, and a growth factor.
  • Examples of a toxin protein (or polypeptide) include, but are not limited to, dipththeria (e.g., diphtheria A chain), Pseudomonas exotoxin and endotoxin, ricin (e.g., ricin A chain), abrin (e.g., abrin A chain), modeccin (e.g., modeccin A chain), alpha-sarcin, Aleurites fordii proteins, dianthin proteins, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtherin toxin, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), Momordica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, mitogellin, restrictocin, phenomycin, enomycin, tricothecenes, inhibitor cystine knot (ICK) peptides (e.g., ceratotoxins), and conotoxin (e.g., KIIIA or Smilla).
  • In some embodiments, the agent is a biocompatible polymer. The BCMA antibodies or the antigen binding fragments as described herein can be conjugated to the biocompatible polymer to increase serum half-life and bioactivity, and/or to extend in vivo half-lives. Examples of biocompatible polymers include water-soluble polymer, such as polyethylene glycol (PEG) or its derivatives thereof and zwitterion-containing biocompatible polymers (e.g., a phosphorylcholine containing polymer).
  • In some embodiments, the agent is an oligonucleotide, such as anti-sense oligonucleotides.
  • In another aspect, the invention provides a conjugate of the antibody or the antigen binding fragment as described herein, wherein the conjugate comprises the formula: antibody-(acyl donor glutamine-containing tag)-(linker)-(cytotoxic agent), wherein the acyl donor glutamine-containing tag is engineered at a specific site of the antibody or the antigen binding fragment (e.g., at a carboxyl terminus of the heavy or light chain, after residue T135 in the antibody heavy chain, or at an another site), wherein the tag is conjugated to a linker (e.g., a linker containing one or more reactive amines (e.g., primary amine NH2)), and wherein the linker is conjugated to a cytotoxic agent (e.g., MMAD or other auristatins such as 0101, 0131, or 3377).
  • Examples of a linker containing one or more reactive amines include, but are not limited to, Ac-Lys-Gly (acetyl-lysine-glycine), aminocaproic acid, Ac-Lys-β-Ala (acetyl-lysine-β-alanine), amino-PEG2 (polyethylene glycol)-C2, amino-PEG3-C2, amino-PEG6-C2 (or amino PEG6-propionyl), Ac-Lys-Val-Cit-PABC (acetyl-lysine-valine-citrulline-p-aminobenzyloxycarbonyl), amino-PEG6-C2-Val-Cit-PABC, aminocaproyl-Val-Cit-PABC, [(3R,5R)-1-{3-[2-(2-aminoethoxy)ethoxy]propanoyl}piperidine-3,5-diyl]bis-Val-Cit-PABC, [(3S,5S)-1-{3-[2-(2-aminoethoxy)ethoxy]propanoyl}piperidine-3,5-diyl]bis-Val-Cit-PABC, putrescine, or Ac-Lys-putrescine.
  • In some embodiments, the conjugate is 1) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101; 2) antibody-AcLys-VC-PABC-0101 and comprises N297Q; 3) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101 and comprises N297Q; 4) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-0131 and comprises N297A; 5) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-3377 and comprises N297A; 6) antibody-GGLLQGA (SEQ ID NO: 475)-AcLys-VC-PABC-0101. In some embodiments, the acyl donor glutamine-containing tag comprising, e.g., GGLLQGPP (SEQ ID NO: 474) or GGLLQGA (SEQ ID NO: 475), is engineered at the C-terminus of the light chain of the antibody. In other embodiments, the acyl donor glutamine-containing tag (e.g., LLQGA (SEQ ID NO: 463) or LLQGPP (SEQ ID NO: 472)) is engineered at the C-terminus of the heavy chain of the antibody, wherein the lysine residue at the C-terminus is deleted. In some embodiments, the acyl donor glutamine-containing tag comprising, e.g., LLQG (SEQ ID NO: 454) is engineered after residue T135 in the antibody heavy chain or replaces amino acid residues E294-N297 in the antibody heavy chain. Examples of the antibody include, but are not limited to, P6E01/P6E01, P6E01/H3.AQ, L1.LGF/L3.KW/P6E01; L1.LGF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.LGF/L3.KW/H3.AL, L1.LGF/L3.KW/H3.AP, L1.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.NY/H3.AL, L1.GDF/L3.NY/H3.AP, L1.GDF/L3.NY/H3.AQ, L3.KW/P6E01, L3.PY/P6E01, L3.NY/P6E01,
      • L3.PY/L1.PS/P6E01, L3.PY/L1.AH/P6E01, L3.PY/L1.FF/P6E01, L3.PY/L1.PH/P6E01, L3.PY/L3.KY/P6E01, L3.PY/L3.KF/P6E01, L3.PY/H2.QR, L3.PY/H2.DY, L3.PY/H2.YQ, L3.PY/H2.LT, L3.PY/H2.HA, L3.PY/H2.QL, L3.PY/H3.YA, L3.PY/H3.AE, L3.PY/H3.AQ, L3.PY/H3.TAQ, L3.PY/P6E01, L3.PY/L1.PS/H2.QR, L3.PY/L1.PS/H2.DY, L3.PY/L1.PS/H2.YQ, L3.PY/L1.PS/H2.LT, L3.PY/L1.PS/H2.HA, L3.PY/L1.PS/H2.QL, L3.PY/L1.PS/H3.YA, L3.PY/L1.PS/H3.AE, L3.PY/L1.PS/H3.AQ, L3.PY/L1.PS/H3.TAQ, L3.PY/L1.AH/H2.QR, L3.PY/L1.AH/H2.DY, L3.PY/L1.AH/H2.YQ, L3.PY/L1.AH/H2.LT, L3.PY/L1.AH/H2.HA, L3.PY/L1.AH/H2.QL, L3.PY/L1.AH/H3.YA, L3.PY/L1.AH/H3.AE, L3.PY/L1.AH/H3.AQ, L3.PY/L1.AH/H3.TAQ, L3.PY/L1.FF/H2.QR, L3.PY/L1.FF/H2.DY, L3.PY/L1.FF/H2.YQ, L3.PY/L1.FF/H2.LT, L3.PY/L1.FF/H2.HA, L3.PY/L1.FF/H2.QL, L3.PY/L1.FF/H3.YA, L3.PY/L1.FF/H3.AE, L3.PY/L1.FF/H3.AQ, L3.PY/L1.FF/H3.TAQ, L3.PY/L1.PH/H2.QR, L3.PY/L1.PH/H2.HA, L3.PY/L1.PH/H3.AE, L3.PY/L1.PH/H3.AQ, L3.PY/L1.PH/H3.TAQ, L3.PY/L3.KY/H2.QR, L3.PY/L3.KY/H2.DY, L3.PY/L3.KY/H2.YQ L3.PY/L3.KY/H2.LT, L3.PY/L3.KY/H2.HA, L3.PY/L3.KY/H2.QL, L3.PY/L3.KY/H3.YA L3.PY/L3.KY/H3.TAQ, L3.PY/L3.KF/H2.DY, L3.PY/L3.KF/H2.YQ, L3.PY/L3.KF/H2.LT L3.PY/L3.KF/H2.QL, L3.PY/L3.KF/H3.YA, L3.PY/L3.KF/H3.AE, L3.PY/L3.KF/H3.AQ L3.PY/L3.KF/H3.TAQ, P5A2_VHVL, A02_Rd4_0.6 nM_C06, A02_Rd4_0.6 nM_C09
      • A02_Rd4_6 nM_C16, A02_Rd4_6 nM_C03, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C26
      • A02_Rd4_6 nM_C25, A02_Rd4_6 nM_C22, A02_Rd4_6 nM_C19, A02_Rd4_0.6 nM_C03
      • A02_Rd4_6 nM_C07, A02_Rd4_6 nM_C23, A02_Rd4_0.6 nM_C18, A02_Rd4_6 nM_C10
      • A02_Rd4_6 nM_C05, A02_Rd4_0.6 nM_C10, A02_Rd4_6 nM_C04, A02_Rd4_0.6 nM_C26
      • A02_Rd4_0.6 nM_C13, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C08, P5C1_VHVL, C01_Rd4_6 nM_C24, C01_Rd4_6 nM_C26, C01_Rd4_6 nM_C10, C01_Rd4_0.6 nM_C27
      • C01_Rd4_6 nM_C20, C01_Rd4_6 nM_C12, C01_Rd4_0.6 nM_C16, C01_Rd4_0.6 nM_C09
      • C01_Rd4_6 nM_C09, C01_Rd4_0.6 nM_C03, C01_Rd4_0.6 nM_C06, C01_Rd4_6 nM_C04
      • COMBO_Rd4_0.6 nM_C22, COMBO_Rd4_6 nM_C21, COMBO_Rd4_6 nM_C10, COMBO_Rd4_0.6 nM_C04, COMBO_Rd4_6 nM_C25, COMBO_Rd4_0.6 nM_C21, COMBO_Rd4_6 nM_C11, COMBO_Rd4_0.6 nM_C20, COMBO_Rd4_6 nM_C09, COMBO_Rd4_6 nM_C08, COMBO_Rd4_0.6 nM_C19, COMBO_Rd4_0.6 nM_C02, COMBO_Rd4_0.6 nM_C23, COMBO_Rd4_0.6 nM_C29, COMBO_Rd4_0.6 nM_C09, COMBO_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C30, COMBO_Rd4_0.6 nM_C14, COMBO_Rd4_6 nM_C07, COMBO_Rd4_6 nM_C02, COMBO_Rd4_0.6 nM_C05, COMBO_Rd4_0.6 nM_C17, COMBO_Rd4_6 nM_C22, COMBO_Rd4_0.6 nM_C11, COMBO_Rd4_0.6 nM_C29, P4G4, or P1A11.
  • In one variation, the conjugate further comprises an amino acid substitution from lysine to arginine at position 222. Accordingly, for example, the conjugate is 1) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101 and comprises K222R; 2) antibody-AcLys-VC-PABC-0101 and comprises N297Q and K222R; 3) antibody-GGLLQGPP (SEQ ID NO: 474)-AcLys-VC-PABC-0101 and comprises N297Q and K222R; 4) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-0131 and comprises N297A and K222R; 5) antibody-LLQG (SEQ ID NO: 454)-amino-PEG6-C2-3377 and comprises N297A and K222R; and 6) antibody-GGLLQGA (SEQ ID NO: 475)-AcLys-VC-PABC-0101 and comprises K222R. In some embodiments, the acyl donor glutamine-containing tag comprising, e.g., GGLLQGPP (SEQ ID NO: 474) or GGLLQGA (SEQ ID NO: 475) is engineered at the C-terminus of the light chain of the antibody. In other embodiments, the acyl donor glutamine-containing tag (e.g., LLQGA (SEQ ID NO: 473) or LLQGPP (SEQ ID NO: 472)) is engineered at the C-terminus of the heavy chain of the antibody, wherein the lysine residue at the C-terminus is deleted. In some embodiments, the acyl donor glutamine-containing tag comprising, e.g., LLQG (SEQ ID NO: 454) is engineered after residue T135 in the antibody heavy chain or replaces amino acid residues E294-N297 in the antibody heavy chain. Examples of the antibody include, but are not limited to, P6E01/P6E01, P6E01/H3.AQ, L1.LGF/L3.KW/P6E01, L1.LGF/L3.NY/P6E01, L1.GDF/L3.NY/P6E01, L1.LGF/L3.KW/H3.AL, L1.LGF/L3.KW/H3.AP, L1.LGF/L3.KW/H3.AQ, L1.LGF/L3.PY/H3.AP, L1.LGF/L3.PY/H3.AQ, L1.LGF/L3.NY/H3.AL, L1.LGF/L3.NY/H3.AP, L1.LGF/L3.NY/H3.AQ, L1.GDF/L3.KW/H3.AL, L1.GDF/L3.KW/H3.AP, L1.GDF/L3.KW/H3.AQ, L1.GDF/L3.PY/H3.AQ, L1.GDF/L3.NY/H3.AL, L1.GDF/L3.NY/H3.AP, L1.GDF/L3.NY/H3.AQ, L3.KW/P6E01, L3.PY/P6E01,
      • L3.NY/P6E01, L3.PY/L1.PS/P6E01, L3.PY/L1.AH/P6E01, L3.PY/L1.FF/P6E01, L3.PY/L1.PH/P6E01, L3.PY/L3.KY/P6E01, L3.PY/L3.KF/P6E01, L3.PY/H2.QR, L3.PY/H2.DY, L3.PY/H2.YQ, L3.PY/H2.LT, L3.PY/H2.HA, L3.PY/H2.QL, L3.PY/H3.YA, L3.PY/H3.AE, L3.PY/H3.AQ, L3.PY/H3.TAQ, L3.PY/P6E01, L3.PY/L1.PS/H2.QR, L3.PY/L1.PS/H2.DY, L3.PY/L1.PS/H2.YQ, L3.PY/L1.PS/H2.LT, L3.PY/L1.PS/H2.HA, L3.PY/L1.PS/H2.QL, L3.PY/L1.PS/H3.YA, L3.PY/L1.PS/H3.AE, L3.PY/L1.PS/H3.AQ, L3.PY/L1.PS/H3.TAQ, L3.PY/L1.AH/H2.QR, L3.PY/L1.AH/H2.DY, L3.PY/L1.AH/H2.YQ, L3.PY/L1.AH/H2.LT, L3.PY/L1.AH/H2.HA, L3.PY/L1.AH/H2.QL, L3.PY/L1.AH/H3.YA, L3.PY/L1.AH/H3.AE, L3.PY/L1.AH/H3.AQ, L3.PY/L1.AH/H3.TAQ, L3.PY/L1.FF/H2.QR, L3.PY/L1.FF/H2.DY, L3.PY/L1.FF/H2.YQ, L3.PY/L1.FF/H2.LT, L3.PY/L1.FF/H2.HA, L3.PY/L1.FF/H2.QL, L3.PY/L1.FF/H3.YA, L3.PY/L1.FF/H3.AE, L3.PY/L1.FF/H3.AQ, L3.PY/L1.FF/H3.TAQ, L3.PY/L1.PH/H2.QR, L3.PY/L1.PH/H2.HA, L3.PY/L1.PH/H3.AE, L3.PY/L1.PH/H3.AQ, L3.PY/L1.PH/H3.TAQ, L3.PY/L3.KY/H2.QR, L3.PY/L3.KY/H2.DY, L3.PY/L3.KY/H2.YQ L3.PY/L3.KY/H2.LT, L3.PY/L3.KY/H2.HA, L3.PY/L3.KY/H2.QL, L3.PY/L3.KY/H3.YA L3.PY/L3.KY/H3.TAQ, L3.PY/L3.KF/H2.DY, L3.PY/L3.KF/H2.YQ, L3.PY/L3.KF/H2.LT L3.PY/L3.KF/H2.QL, L3.PY/L3.KF/H3.YA, L3.PY/L3.KF/H3.AE, L3.PY/L3.KF/H3.AQ L3.PY/L3.KF/H3.TAQ, P5A2_VHVL, A02_Rd4_0.6 nM_C06, A02_Rd4_0.6 nM_C09
      • A02_Rd4_6 nM_C16. A02_Rd4_6 nM C03. A02_Rd4_6 nM_C01. A02_Rd4_6 nM_C26
      • A02_Rd4_6 nM_C25, A02_Rd4_6 nM_C22, A02_Rd4_6 nM_C19, A02_Rd4_0.6 nM_C03
      • A02_Rd4_6 nM_C07, A02_Rd4_6 nM_C23, A02_Rd4_0.6 nM_C18, A02_Rd4_6 nM_C10
      • A02_Rd4_6 nM_C05, A02_Rd4_0.6 nM_C10, A02_Rd4_6 nM_C04, A02_Rd4_0.6 nM_C26
      • A02_Rd4_0.6 nM_C13, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C08, P5C1_VHVL, C01_Rd4_6 nM_C24, C01_Rd4_6 nM_C26, C01_Rd4_6 nM_C10, C01_Rd4_0.6 nM_C27
      • C01_Rd4_6 nM_C20, C01_Rd4_6 nM_C12, C01_Rd4_0.6 nM_C16, C01_Rd4_0.6 nM_C09
      • C01_Rd4_6 nM_C09, C01_Rd4_0.6 nM_C03, C01_Rd4_0.6 nM_C06, C01_Rd4_6 nM_C04
      • COMBO_Rd4_0.6 nM_C22, COMBO_Rd4_6 nM_C21, COMBO_Rd4_6 nM_C10, COMBO_Rd4_0.6 nM_C04, COMBO_Rd4_6 nM_C25, COMBO_Rd4_0.6 nM_C21, COMBO_Rd4_6 nM_C11, COMBO_Rd4_0.6 nM_C20, COMBO_Rd4_6 nM_C09, COMBO_Rd4_6 nM_C08, COMBO_Rd4_0.6 nM_C19, COMBO_Rd4_0.6 nM_C02, COMBO_Rd4_0.6 nM_C23, COMBO_Rd4_0.6 nM_C29, COMBO_Rd4_0.6 nM_C09, COMBO_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C30, COMBO_Rd4_0.6 nM_C14, COMBO_Rd4_6 nM_C07, COMBO_Rd4_6 nM_C02, COMBO_Rd4_0.6 nM_C05, COMBO_Rd4_0.6 nM_C17, COMBO_Rd4_6 nM_C22, COMBO_Rd4_0.6 nM_C11, COMBO_Rd4_0.6 nM_C29, or P4G4, or P1A11.
    CD3 Antibodies and Methods of Making Thereof
  • The present invention further provides an antibody that binds to CD3 (e.g., human CD3 (SEQ ID NO: 502; or accession number: NM_000733.3).
  • In one aspect, provided is an isolated antibody, or an antigen binding fragment thereof, which specifically binds to CD3, wherein the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443, 445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399.
  • In another aspect, provided is an isolated antibody, or an antigen binding fragment thereof, which specifically binds to CD3, wherein the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 340, 343, 430, 431, 435, or 440, 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341, 433, 452, or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342, 432, 434, 437, 438, 439, 446, or 453.
  • In some embodiments, provided is an antibody having any one of partial light chain sequence as listed in Table 3 and/or any one of partial heavy chain sequence as listed in Table 3.
  • TABLE 3
    mAb Light Chain Heavy Chain
    h2B4 DIVMTQSPDSLAVSLGERATINC T EVQLVESGGGLVQPGGSLRLSCA
    SSQSLFNVRSRKNYLA WYQQKP ASGFTFS DYYMTWVRQAPGKGLE
    GQPPKLLIS WASTRES GVPDRFS WVAFIRNRARGYTSDHNASVKGR
    GSGSGTDFTLTISSLQAEDVAVY FTISRDNAKNSLYLQMNSLRAEDT
    YC KQSYDLFT FGSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 319) VTVSS
    (SEQ ID NO: 320)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH-wt KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    VL_TK PGQPPKLLIS WASTRES GVPDRF WVAFIRNRARGYTSDHNASVKGR
    SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    YYC KQSYDLFT FGSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 321) VTVSS
    (SEQ ID NO: 322)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH- KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    hnps PGQPPKLLIS WASTRES GVPDRF WAFIRNRARGYTSDHNPSVKGR
    VL_TK SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    YYC KQSYDLFT FGSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 323) VTVSS
    (SEQ ID NO: 324)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH- KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    yaes PGQPPKLLIS WASTRES GVPDRF WAFIRNRARGYTSDYAESVKGR
    VL_TK SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    YYC KQSYDLFTF GSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 325) VTVSS
    (SEQ ID NO: 326)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH- KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    yads PGQPPKLLIS WASTRES GVPDRF WVAFIRNRARGYTSDYADSVKGR
    VL_TK SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    YYC KQSYDLFT FGSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 327) VTVSS
    (SEQ ID NO: 328)
    h2B4- DIVMTQSPDSLAVSLGERATINC T EVQLVESGGGLVQPGGSLRLSCA
    VH- SSQSLFNVRSRKNYLA WYQQKP ASGFTFS DYYMTWVRQAPGKGLE
    yaps GQPPKLLIS WASTRES GVPDRFS WVAFIRNRARGYTSDYAPSVKGR
    VL_TK GSGSGTDFTLTISSLQAEDVAVY FTISRDNAKNSLYLQMNSLRAEDT
    YC KQSYDLFTF GSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 329) VTVSS
    (SEQ ID NO: 330)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH- KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    hnps PGQPPKLLIY WASTRES GVPDRF WVAFIRNRARGYTSDHNPSVKGR
    VL_TK- SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    S55Y YYC KQSYDLFT FGSGTKLEIK AVYYCARD RPSYYVLDY WGQGTT
    (SEQ ID NO: 344) VTVSS (SEQ ID NO: 345)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH- KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    hnps PGQPPKLLIS WASTRES GVPDRF WVAFIRNRARGYTSDHNPSVKGR
    VL_TK- SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    S105Q YYC KQSYDLFT FGQGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 346) VTVSS (SEQ ID NO: 347)
    h2B4- DIVMTQSPDSLAVSLGERATINC EVQLVESGGGLVQPGGSLRLSCA
    VH- KSSQSLFNVRSRKNYLA WYQQK ASGFTFS DYYMTWVRQAPGKGLE
    hnps PGQPPKLLIY WASTRES GVPDRF WVAFIRNRARGYTSDHNPSVKGR
    VL_TK- SGSGSGTDFTLTISSLQAEDVAV FTISRDNAKNSLYLQMNSLRAEDT
    S55Y/ YYC KQSYDLFT FGQGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    S105Q (SEQ ID NO: 348) VTVSS (SEQ ID NO: 349)
    2B4 DIVMSQSPPSLAVSVGDKVTMSC EVKLVESGGGLVQPGGSLRLSCA
    TSSQSLFNSRSRKNYLA WYQQK TFGFTFT DYYMTWVRQPPGKALE
    SGQSPKLLIS WASTRES GVPDRF WVAFIRNRARGYTSDHNASVKGR
    TGSGSGTDFTLTISSVQAEDLAV FTISRDNSQNILYLQMNTLRAEDS
    YYC KQSYDLFT FGSGTKLEIK ATYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 350) VTVSS (SEQ ID NO: 351)
    h2B4- DIVMTQSPDSLAVSLGERATINC T EVQLVESGGGLVQPGGSLRLSCA
    11 SSQSLFNSRSRKNYLA WYQQKP ASGFTFS DYYMTWVRQAPGKGLE
    GQPPKLLIS WASTRES GVPDRFS WVAFIRNRARGYTSDHNASVKGR
    GSGSGTDFTLTISSLQAEDVAVY FTISRDNAKNSLYLQMNSLRAEDT
    YC QQSYDTFT FGSGTKLEIK AVYYCAR DRPSYYVLDY WGQGTT
    (SEQ ID NO: 445) VTVSS (SEQ ID NO: 444)
    1C10 DIVMSQSPSSLAVSAGEKVTMSC QVQLQQPGSELVRPGASVILSCKA
    KSSQSLLNSRTRKNYLA WYQQK SGYTFT SYWMHWVRQRPGQGLE
    PGQSPKLLIY WASTRES GVPDRF WIGNIYSGGDTINYDEKFKNKAILT
    TGSGSGTDFTLTIDSVQPEDLAV VDTSSSTAYMHLSSLTSEDSAVYY
    YYC TQSFILRT FGGGTKLEIK CTR DATSRYFFDY WGQGTTVTVS
    (SEQ ID NO: 352) S (SEQ ID NO: 354)
    1A4 DIVMSQSPSSLAVSAGEKVTMSC QVQLQQSGPDLVKPGASVEISCK
    KSSQSLLNSRTRKNYLA WYQQK ASGYSFT TYYLHWVRQRPGQGLE
    PGQSPKLLIY WASTRAS GVPDRF WIGWIFPGSDNTKYNEKFKGKATL
    TGSGSGTDFTLTISSVQAEDLAIY TADTSSSTAYMQLSSLTSEDSAVY
    Y CKQSFILRT FGGGTKLEIK (SEQ FCAR NRDYYFDY WGQGTTVTVSS
    ID NO: 355) (SEQ ID NO: 356)
    7A3 DIVVSQSPSSLAVSAGEKVIMSC EVQLQQSGAELVRPGALVKLSCK
    KSSQSLLNSRTRKNYLA WYQLK GSGFNIK DYYIHWVKQRPEQGLE
    PGQSPKLLIY SASTRES GVPDRF WIGWIDPENGNNKYDPKFQGKASI
    TGSGSGTDFTLTISSVQTEDLAV TADTSSNIAYLQLSSLTSEDTAVYY
    YYC MQSFTLRT FGGGTKLEIK CAR NDNYAFDY WGQGTTVTVSS
    (SEQ ID NO: 443) (SEQ ID NO: 442)
    25A8 QAVVTQESALTTSPGEAVTLTC R EVQLVESGGGLVRPEGSLRLSCA
    SSTGAVTTSNYAN WVQEKPDHL ASGFTFNTYAMNWVRQAPGKGLE
    FTGLIG GTNTRAP GVPARFSGSLI WVGRIRSKINNYATYYAESVKGRF
    GDKAALTITGAQTEDEAIYFC VL TLSRDDSLSMVYLQMNSLKNEDT
    WYNNYWV FGGGTKLTVL (SEQ AMYYCVR HETLRSGISWFAS WGQ
    ID NO: 377) GTLVTVSS (SEQ ID NO: 378)
    16G7 QAVVTQESALTTSPGETVTLTC R EVQLVDSGGGLVQPKGSLKLSCA
    SSTGAVTTSNYAN WVQEKPDHL ASGFTFNTYAMNWVRQAPGKGLE
    FTGLIG GTNNRAP GVPARFSGSL WVARIRSKSNNYATYYADSVKDR
    IGDKAALTITGAQTEDEAIYFC AL FTISRDDSQSRLYLQMNNLKTEDT
    WYSNHWV FGGGTKLTVL (SEQ AMYYCVR HETLRSGISWFAN WG
    ID NO: 379) QGTLVTVSS (SEQ ID NO: 380)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    B5 SSTGAVTTSNYAN WVQQKPGQA ASGFTFSTYAMNWVRQAPGKGLE
    PRGLIG GTNTRAP GTPARFSGSL WVGRIRSKINNYATYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNYWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 381) TLVTVSS (SEQ ID NO: 382)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    B8 SSTGAVTTSNYAN WVQQKPGQA ASGFTFSTYAMNWVRQAPGKGLE
    PRGLIG GTNTRAP GTPARFSGSL WVGRIRSKINNYATYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNHWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 383) TLVTVSS (SEQ ID NO: 384)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    B12 ASTGAVTTSNYAN WVQQKPGQ ASGFTFSTYAMNWVRQAPGKGLE
    APRGLIG GTNTRAP GTPARFSGS WVGRIRSKINNYATYYAESVKGRF
    LLGGKAALTLSGAQPEDEAEYYC TISRDDSKNTLYLQMNSLKTEDTA
    VLWYNNHWV FGGGTKLTVL VYYCVRH ETLRSGISWFAS WGQG
    (SEQ ID NO: 385) TLVTVSS (SEQ ID NO: 386)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    B13 TSTGAVTTSNYAN WVQQKPGQA ASGFTFSTYAMNWVRQAPGKGLE
    PRGLIGGTNTRAPGTPARFSGSL WVGRIRSKINNYATYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNHWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 387) TLVTVSS (SEQ ID NO: 388)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    C5 SSTGAVTTSNYAN WVQQKPGQA ASGFTFN TYAMNWVRQAPGKGLE
    PRGLIG GTNTRAP GTPARFSGSL WVGRIRSKINNYATYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNYWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 389) TLVTVSS (SEQ ID NO: 390)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    C8 SSTGAVTTSNYAN WVQQKPGQA ASGFTFN TYAMNWVRQAPGKGLE
    PRGLIG GTNTRAP GTPARFSGSL WVGRIRSKINNYATYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNHWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 391) TLVTVSS (SEQ ID NO: 392)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    D13 TSTGAVTTSNYAN WVQQKPGQA ASGFTFS TYAMNWVRQAPGKGLE
    PRGLIGGTNTRAPGTPARFSGSL WVGRIRSHINNYATYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNHWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 393) TLVTVSS (SEQ ID NO: 394)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    E13 TSTGAVTTSNYAN WVQQKPGQA ASGFTFS TYAMNWVRQAPGKGLE
    PRGLIGGTNTRAPGTPARFSGSL WVGRIRSKYNNYATYYAESVKGR
    LGGKAALTLSGAQPEDEAEYYC V FTISRDDSKNTLYLQMNSLKTEDT
    LWYNNHWV FGGGTKLTVL (SEQ AVYYCVR HETLRSGISWFAS WGQ
    ID NO: 395) GTLVTVSS (SEQ ID NO: 396)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    F13 TSTGAVTTSNYAN WVQQKPGQA ASGFTFS TYAMNWVRQAPGKGLE
    PRGLIGGTNTRAPGTPARFSGSL WVGRERSKINNYATYYAESVKGR
    LGGKAALTLSGAQPEDEAEYYC V FTISRDDSKNTLYLQMNSLKTEDT
    LWYNNHWV FGGGTKLTVL (SEQ AVYYCVR HETLRSGISWFAS WGQ
    ID NO: 397) GTLVTVSS (SEQ ID NO: 398)
    h25A8- QAVVTQEPSLTVSPGGTVTLTC R EVQLVESGGGLVKPGGSLRLSCA
    G13 TSTGAVTTSNYAN WVQQKPGQA ASGFTFS TYAMNWVRQAPGKGLE
    PRGLIGGTNTRAPGTPARFSGSL WVGRIRSKINNYKTYYAESVKGRF
    LGGKAALTLSGAQPEDEAEYYC V TISRDDSKNTLYLQMNSLKTEDTA
    LWYNNHWV FGGGTKLTVL (SEQ VYYCVR HETLRSGISWFAS WGQG
    ID NO: 399) TLVTVSS (SEQ ID NO: 400)

    In Table 3, the underlined sequences are CDR sequences according to Kabat and in bold according to Chothia.
  • The invention also provides CDR portions of antibodies to CD3 (including Chothia, Kabat CDRs, and CDR contact regions). Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed “combined CRs” or “extended CDRs”). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof. Table 4 provides examples of CDR sequences provided herein.
  • TABLE 4
    Heavy Chain
    mAb CDRH1 CDRH2 CDRH3
    h2B4 DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    GFTFSDY (SEQ ID FIRNRARGYTSDHNASVKG
    NO: 332) (Chothia); (SEQ ID NO: 334) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-wt 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK GFTFSDY (SEQ ID FIRNRARGYTSDHNASVKG
    NO: 332) (Chothia); (SEQ ID NO: 334) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-hnps 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK GFTFSDY (SEQ ID FIRNRARGYTSDHNPSVKG
    NO: 332) (Chothia); (SEQ ID NO: 336) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-yaes 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK GFTFSDY (SEQ ID FIRNRARGYTSDYAESVKG
    NO: 332) (Chothia); (SEQ ID NO: 337) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-yads 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK GFTFSDY (SEQ ID FIRNRARGYTSDYADSVKG
    NO: 332) (Chothia); (SEQ ID NO: 338) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-yaps 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK GFTFSDY (SEQ ID FIRNRARGYTSDYAPSVKG
    NO: 332) (Chothia); (SEQ ID NO: 339) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-hnps 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK- GFTFSDY (SEQ ID FIRNRARGYTSDHNPSVKG
    S55Y NO: 332) (Chothia); (SEQ ID NO: 336) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-hnps 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK- GFTFSDY (SEQ ID FIRNRARGYTSDHNPSVKG
    S105Q NO: 332) (Chothia); (SEQ ID NO: 336) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    h2B4- DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    VH-hnps 331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    VL_TK- GFTFSDY (SEQ ID FIRNRARGYTSDHNPSVKG
    S55Y/ NO: 332) (Chothia); (SEQ ID NO: 336) (Kabat)
    S105Q GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    2B4 DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    GFTFTDY (SEQ ID FIRNRARGYTSDHNASVKG
    NO: 415) (Chothia); (SEQ ID NO: 418) (Kabat)
    GFTFTDYYMT (SEQ
    ID NO: 416)
    (Extended)
    h2B4-11 DYYMT (SEQ ID NO: RNRARGYT DRPSYYVLDY
    331) (Kabat); (SEQ ID NO: 417) (Chothia) (SEQ ID NO: 335)
    GFTFSDY (SEQ ID FIRNRARGYTSDHNASVKG
    NO: 332) (Chothia); (SEQ ID NO: 418) (Kabat)
    GFTFSDYYMT (SEQ
    ID NO: 333)
    (Extended)
    1C10 SYWMH (SEQ ID NO: YSGGDT DATSRYFFDY
    418) (Kabat) (SEQ ID NO: 421) (Chothia) (SEQ ID NO: 423)
    GYTFTSY (SEQ ID NIYSGGDTINYDEKFKN
    NO: 419) (Chothia) (SEQ ID NO: 422) (Kabat)
    GYTFTSYWMH (SEQ
    ID NO: 420)
    (Extended)
    1A4 TYYLH (SEQ ID NO: FPGSDN (SEQ ID NO: 427) NRDYYFDY
    424) (Kabat) (Chothia) (SEQ ID NO: 429)
    GYSFTTYY (SEQ ID WIFPGSDNTKYNEKFKG
    NO: 425) (Chothia) (SEQ ID NO: 428) (Kabat)
    GYSFTTYYLH (SEQ
    ID NO: 426)
    (Extended)
    7A3 DYYIH (SEQ ID NO: DPENGN (SEQ ID NO: 449) NDNYAFDY
    446) (Kabat) (Chothia) (SEQ ID NO: 451)
    GFNIKDY(SEQ ID WIDPENGNNKYDPKFQG
    NO: 447) (Chothia) (SEQ ID NO: 450) (Kabat)
    GFNIKDYYIH (SEQ
    ID NO: 448)
    (Extended)
    25A8 TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFNTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 402) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFNTYAMN (SEQ
    ID NO: 403)
    (Extended)
    16G7 TYAMN (SEQ ID NO: RSKSNNYA (SEQ ID NO: HETLRSGISWFA
    401) (Kabat); 404) (Chothia) N (SEQ ID NO:
    GFTFNTY (SEQ ID RIRSKSNNYATYYADSVKD 406)
    NO: 402) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFNTYAMN (SEQ
    ID NO: 403)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    B5 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    B8 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    B12 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    B13 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    C5 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFNTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 402) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFNTYAMN (SEQ
    ID NO: 403)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    C8 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFNTY (SEQ ID RIRSKINNYATYYAESVKG 406)
    NO: 402) (Chothia); (SEQ ID NO: 405) (Kabat)
    GFTFNTYAMN (SEQ
    ID NO: 403)
    h25A8- TYAMN (SEQ ID NO: RSHINNYA (SEQ ID NO: HETLRSGISWFA
    D13 401) (Kabat); 409) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSHINNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 410) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKYNNYA (SEQ ID NO: HETLRSGISWFA
    E13 401) (Kabat); 411) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSKYNNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 412) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    F13 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RERSKINNYATYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 413) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    h25A8- TYAMN (SEQ ID NO: RSKINNYA (SEQ ID NO: HETLRSGISWFA
    G13 401) (Kabat); 404) (Chothia) S (SEQ ID NO:
    GFTFSTY (SEQ ID RIRSKINNYKTYYAESVKG 406)
    NO: 407) (Chothia); (SEQ ID NO: 414) (Kabat)
    GFTFSTYAMN (SEQ
    ID NO: 408)
    (Extended)
    Light Chain
    mAb CDRH1 CDRH2 CDRH3
    h2B4 TSSQSLFNVRSRKN WASTRES KQSYDLFT
    YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    (SEQ ID NO: 340)
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-wt YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK (SEQ ID NO: 343)
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-hnps YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK (SEQ ID NO: 343)
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-yaes YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK (SEQ ID NO: 343)
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-yads YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK (SEQ ID NO: 343)
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-yaps YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK (SEQ ID NO: 343)
    2B4 TSSQSLFNSRSRKN WASTRES KQSYDLFT
    YLA (SEQ ID NO: (SEQ ID NO: 341) (SEQ ID NO: 342)
    430)
    h2B4-11 TSSQSLFNSRSRKN WASTRES QQSYDTFT (SEQ
    YLA (SEQ ID NO: (SEQ ID NO: 341) ID NO: 446)
    430)
    1C10 KSSQSLLNSRTRKN WASTRES TQSFILRT (SEQ
    Y (SEQ ID NO: 431) (SEQ ID NO: 341) ID NO: 432)
    1A4 KSSQSLLNSRTRKN WASTRAS KQSFILRT (SEQ
    Y (SEQ ID NO: 431) (SEQ ID NO: 433) ID NO: 434)
    7A3 KSSQSLLNSRTRKN SASTRES MQSFTLRT (SEQ
    Y (SEQ ID NO: 431) (SEQ ID NO: 452) ID NO: 453)
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-hnps YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK- (SEQ ID NO: 343)
    S55Y
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-hnps YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK- (SEQ ID NO: 343)
    S105Q
    h2B4- KSSQSLFNVRSRKN WASTRES KQSYDLFT
    VH-hnps YLA (SEQ ID NO: 341) (SEQ ID NO: 342)
    VL_TK- (SEQ ID NO: 343)
    S55Y/
    S105Q
    25A8 RSSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNYWV
    (SEQ ID NO: 435) (SEQ ID NO: 437)
    16G7 RSSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) ALWYSNHWV
    (SEQ ID NO: 435) (SEQ ID NO: 438)
    h25A8- RSSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNYWV
    B5 (SEQ ID NO: 435) (SEQ ID NO: 437)
    h25A8- RSSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    B8 (SEQ ID NO: 435) (SEQ ID NO: 439)
    h25A8- RASTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    B12 (SEQ ID NO: 440) (SEQ ID NO: 439)
    h25A8- RTSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    B13 (SEQ ID NO: 441) (SEQ ID NO: 439)
    h25A8- RSSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNYWV
    C5 (SEQ ID NO: 435) (SEQ ID NO: 437)
    h25A8- RSSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    C8 (SEQ ID NO: 435) (SEQ ID NO: 439)
    h25A8- RTSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    D13 (SEQ ID NO: 441) (SEQ ID NO: 439)
    h25A8- RTSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    E13 (SEQ ID NO: 441) (SEQ ID NO: 439)
    h25A8- RTSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    F13 (SEQ ID NO: 441) (SEQ ID NO: 439)
    h25A8- RTSTGAVTTSNYAN GTNTRAP (SEQ ID NO: 436) VLWYNNHWV
    G13 (SEQ ID NO: 441) (SEQ ID NO: 439)
  • The invention also provides isolated polynucleotides encoding the antibodies of the invention, and vectors and host cells comprising the polynucleotide.
  • In one embodiment, a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody h2B4, h2B4-VH-wt VL_TK, h2B4-VH-hnps VL_TK, h2B4-VH-yaes VL_TK, h2B4-VH-yads VL_TK, h2B4-VH-yaps VL_TK, h2B4-VH-hnps VL_TK-S55Y, h2B4-VH-hnps VL_TK-S105Q, h2B4-vH-hnps VL_TK-S55Y/S105Q, 2B34, h21B4-11, 1010, 1A4, 7A3, 25A8, 16G7, h25A8-B5, h25A8-B8, h25A8-B12, h25A8-B13, h25A8-C5, h25A8-C8, h25A8-D13, h25A8-E13, h25A8-F13, or h25A8-G13. The sequence encoding the antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use. Vectors (including expression vectors) and host cells are further described herein.
  • The invention also encompasses fusion proteins comprising one or more fragments or regions from the antibodies of this invention. In one embodiment, a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of the variable light chain region shown in SEQ ID NOs: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 445, 352, 355, 443, 377, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399, and/or at least 10 amino acids of the variable heavy chain region shown in SEQ ID NOs: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 354, 356, 444, 442, 378, 380, 382, 384, 386, 388, 390, 392, 394, 396, 398, or 400. In other embodiments, a fusion polypeptide is provided that comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region. In another embodiment, the fusion polypeptide comprises a light chain variable region and/or a heavy chain variable region, as shown in any of the sequence pairs selected from among SEQ ID NOs: 319 and 320, 321 and 322, 323 and 324, 325 and 326, 327 and 328, 329 and 330, 344 and 345, 346 and 347, 348 and 349, 350 and 351, 445 and 444, 352 and 354, 355 and 356, 443 and 442, 377 and 378, 379 and 380, 381 and 382, 383 and 384, 385 and 386, 387 and 388, 389 and 390, 391 and 392, 393 and 394, 395 and 396, 397 and 398, or 399 and 400. In another embodiment, the fusion polypeptide comprises one or more CDR(s). In still other embodiments, the fusion polypeptide comprises CDR H3 (VH CDR3) and/or CDR L3 (VL CDR3). For purposes of this invention, a fusion protein contains one or more antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region. Exemplary heterologous sequences include, but are not limited to a “tag” such as a FLAG tag or a 6His tag. Tags are well known in the art.
  • A fusion polypeptide can be created by methods known in the art, for example, synthetically or recombinantly. Typically, the fusion proteins of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, although they may also be prepared by other means known in the art, including, for example, chemical synthesis.
  • Representative materials of the CD3 antibody in the present invention were deposited in the American Type Culture Collection (ATCC) on Sep. 11, 2015. Vector having ATCC Accession No. PTA-122513 is a polynucleotide encoding a humanized CD3 antibody heavy chain variable region, and vector having ATCC Accession No. PTA-122512 is a polynucleotide encoding a humanized CD3 antibody light chain variable region. The deposits were made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and Regulations thereunder (Budapest Treaty). This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit. The deposit will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Pfizer, Inc. and ATCC, which assures permanent and unrestricted availability of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U.S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 U.S.C. Section 122 and the Commissioner's rules pursuant thereto (including 37 C.F.R. Section 1.14 with particular reference to 886 OG 638).
  • The assignee of the present application has agreed that if a culture of the materials on deposit should die or be lost or destroyed when cultivated under suitable conditions, the materials will be promptly replaced on notification with another of the same. Availability of the deposited material is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws.
  • Bispecific Antibodies and Methods of Making
  • Bispecific antibodies, monoclonal antibodies that have binding specificities for at least two different antigens, can be prepared using the antibodies disclosed herein. Methods for making bispecific antibodies are known in the art (see, e.g., Suresh et al., Methods in Enzymology 121:210, 1986). Traditionally, the recombinant production of bispecific antibodies was based on the coexpression of two immunoglobulin heavy chain-light chain pairs, with the two heavy chains having different specificities (Millstein and Cuello, Nature 305, 537-539, 1983).
  • According to one approach to making bispecific antibodies, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant region sequences. The fusion preferably is with an immunoglobulin heavy chain constant region, comprising at least part of the hinge, CH2 and CH3 regions. It is preferred to have the first heavy chain constant region (CH1), containing the site necessary for light chain binding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are cotransfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance.
  • In one approach, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. This asymmetric structure, with an immunoglobulin light chain in only one half of the bispecific molecule, facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations. This approach is described in PCT Publication No. WO 94/04690.
  • In another approach, the bispecific antibodies are composed of amino acid modification in the first hinge region in one arm, and the substituted/replaced amino acid in the first hinge region has an opposite charge to the corresponding amino acid in the second hinge region in another arm. This approach is described in International Patent Application No. PCT/US2011/036419 (WO2011/143545).
  • In another approach, the formation of a desired heteromultimeric or heterodimeric protein (e.g., bispecific antibody) is enhanced by altering or engineering an interface between a first and a second immunoglobulin-like Fc region (e.g., a hinge region and/or a CH3 region). In this approach, the bispecific antibodies may be composed of a CH3 region, wherein the CH3 region comprises a first CH3 polypeptide and a second CH3 polypeptide which interact together to form a CH3 interface, wherein one or more amino acids within the CH3 interface destabilize homodimer formation and are not electrostatically unfavorable to homodimer formation. This approach is described in International Patent Application No. PCT/US2011/036419 (WO2011/143545).
  • In another approach, the bispecific antibodies can be generated using a glutamine-containing peptide tag engineered to the antibody directed to an epitope (e.g., BCMA) in one arm and another peptide tag (e.g., a Lys-containing peptide tag or a reactive endogenous Lys) engineered to a second antibody directed to a second epitope in another arm in the presence of transglutaminase. This approach is described in International Patent Application No. PCT/IB2011/054899 (WO2012/059882).
  • In another aspect of the invention, the heterodimeric protein (e.g., bispecific antibody) as described herein comprises a full-length human antibody, wherein a first antibody variable domain of the heterodimeric protein is capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen located on the human immune effector cell, and wherein a second antibody variable domain of the heterodimeric protein is capable of specifically binding to a target antigen. In some embodiments, the human antibody has an IgG1, IgG2, IgG3, or IgG4 isotype. In some embodiments, the heterodimeric protein comprises an immunologically inert Fc region.
  • The human immune effector cell can be any of a variety of immune effector cells known in the art. For example, the immune effector cell can be a member of the human lymphoid cell lineage, including, but not limited to, a T cell (e.g., a cytotoxic T cell), a B cell, and a natural killer (NK) cell. The immune effector cell can also be, for example without limitation, a member of the human myeloid lineage, including, but not limited to, a monocyte, a neutrophilic granulocyte, and a dendritic cell. Such immune effector cells may have either a cytotoxic or an apoptotic effect on a target cell or other desired effect upon activation by binding of an effector antigen.
  • The effector antigen is an antigen (e.g., a protein or a polypeptide) that is expressed on the human immune effector cell. Examples of effector antigens that can be bound by the heterodimeric protein (e.g., a heterodimeric antibody or a bispecific antibody) include, but are not limited to, human CD3 (or CD3 (Cluster of Differentiation) complex), CD16, NKG2D, NKp46, CD2, CD28, CD25, CD64, and CD89.
  • The target cell can be a cell that is native or foreign to humans. In a native target cell, the cell may have been transformed to be a malignant cell or pathologically modified (e.g., a native target cell infected with a virus, a plasmodium, or a bacterium). In a foreign target cell, the cell is an invading pathogen, such as a bacterium, a plasmodium, or a virus.
  • The target antigen is expressed on a target cell in a diseased condition (e.g., an inflammatory disease, a proliferative disease (e.g., cancer), an immunological disorder, a neurological disease, a neurodegenerative disease, an autoimmune disease, an infectious disease (e.g., a viral infection or a parasitic infection), an allergic reaction, a graft-versus-host disease or a host-versus-graft disease). A target antigen is not effector antigen. Examples of the target antigens include, but are not limited to, BCMA, EpCAM (Epithelial Cell Adhesion Molecule), CCR5 (Chemokine Receptor type 5), CD19, HER (Human Epidermal Growth Factor Receptor)-2/neu, HER-3, HER-4, EGFR (Epidermal Growth Factor Receptor), PSMA, CEA, MUC-1 (Mucin), MUC2, MUC3, MUC4, MUCSAC, MUC5B, MUC7, ClhCG, Lewis-Y, CD20, CD33, CD30, ganglioside GD3, 9-O-Acetyl-GD3, GM2, Globo H, fucosyl GM1, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Shh (Sonic Hedgehog), Wue-1, Plasma Cell Antigen, (membrane-bound) IgE, MCSP (Melanoma Chondroitin Sulfate Proteoglycan), CCR8, TNF-alpha precursor, STEAP, mesothelin, A33 Antigen, PSCA (Prostate Stem Cell Antigen), Ly-6; desmoglein 4, E-cadherin neoepitope, Fetal Acetylcholine Receptor, CD25, CA19-9 marker, CA-125 marker and MIS (Muellerian Inhibitory Substance) Receptor type II, sTn (sialylated Tn antigen; TAG-72), FAP (fibroblast activation antigen), endosialin, EGFRvIII, LG, SAS and CD63.
  • In some embodiments, the heterodimeric protein (e.g., bispecific antibody) as described herein comprises a full-length human antibody, wherein a first antibody variable domain of the heterodimeric protein is capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen (e.g., CD3 antigen) located on the human immune effector cell, wherein a second antibody variable domain of the heterodimeric protein is capable of specifically binding to a target antigen (e.g., CD20 antigen or EpCAM), wherein the first and second antibody variable domain of the heterodimeric protein comprise amino acid modifications at positions 223, 225, and 228 (e.g., (C223E or C223R), (E225R), and (P228E or P228R)) in the hinge region and at position 409 or 368 (e.g., K409R or L368E (EU numbering scheme)) in the CH3 region of human IgG2 (SEQ ID NO: 493).
  • In some embodiments, the first and second antibody variable domains of the heterodimeric protein comprise amino acid modifications at positions 221 and 228 (e.g., (D221R or D221E) and (P228R or P228E)) in the hinge region and at position 409 or 368 (e.g., K409R or L368E (EU numbering scheme)) in the CH3 region of human IgG1 (SEQ ID NO: 494).
  • In some embodiments, the first and second antibody variable domains of the heterodimeric protein comprise amino acid modifications at positions 228 (e.g., (P228E or P228R)) in the hinge region and at position 409 or 368 (e.g., R409 or L368E (EU numbering scheme)) in the CH3 region of human IgG4 (SEQ ID NO: 495).
  • In another embodiment, the first antibody variable domain of the heterodimeric protein comprises a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443, 445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399, and the second antibody variable domain of the heterodimeric protein comprises VH region comprising the VH sequence shown in SEQ ID NO: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 125, 127, 313, 314, 363, or 365; and/or a VL region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38, 40, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 317, 81, 82, 84, 85, 86, 88, 89, 90, 91, 93, 94, 96, 98, 100, 102, 103, 105, 107, 108, 109, 111, 113, 115, 116, 117, 119, 121, 123, 124, 126, 128, 315, or 364.
  • In another embodiment, the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387; and the second antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 112; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 38.
  • In another embodiment, the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 417; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 156; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 159; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 225. The VH CDR1 of the first antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 331. For example, the VH CDR1 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 333. The VH CDR2 of the first antibody variable domain may further comprise the sequence of F or FI immediately preceding the sequence shown in SEQ ID NO: 417 and/or the sequence of S, SD, SDH, SDHN, SDHNP, SDHNPS, SDHNPSV, SDHNPSVK, or SDHNPSVKG immediately following the sequence shown in SEQ ID NO: 417. For example, the VH CDR2 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 336. The VH CDR1 of the second antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 156. For example, the VH CDR1 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 157. The VH CDR2 of the second antibody variable domain may further comprise the sequence of A or AI immediately preceding the sequence shown in SEQ ID NO: 159 and/or the sequence of L, LP, LPY, LPYA, LPYAD, LPYADS, LPYADSV, LPYADSVK, or LPYADSVKG immediately following the sequence shown in SEQ ID NO: 159. For example, the VH CDR2 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 158.
  • In another embodiment, the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 332 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 151; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 158; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 225. The VH CDR1 of the first antibody variable domain may further comprise the sequence of Y, YM or YMT immediately following the sequence shown in SEQ ID NO: 332. For example, the VH CDR1 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 333. The VH CDR1 of the second antibody variable domain may further comprise the sequence P, PM, or PMS immediately following the sequence shown in SEQ ID NO: 151. For example, the VH CDR1 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 157.
  • In another embodiment, the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 401 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 404; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 406; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 439; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 156; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 159; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 225. The VH CDR1 of the first antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 401. For example, the VH CDR1 of the first antibody variable domain comprises the sequence shown in SEQ ID NO: 408. The VH CDR2 of the first antibody variable domain may further comprise the sequence of R or RI immediately preceding the sequence shown in SEQ ID NO: 404 and/or the sequence of T, TY, TYY, TYYA, TYYAE, TYYAES, TYYAESV, TYYAESVK, or TYYAESVKG immediately following the sequence shown in SEQ ID NO: 404. For example, the bispecific antibody of claim 49, wherein the VH CDR2 of the first antibody variable domain comprises the sequence shown in SEQ ID NO: 405. The VH CDR1 of the second antibody variable domain may further comprise the sequence of S, FS, TFS, FTFS or GFTFS immediately preceding the sequence shown in SEQ ID NO: 156. For example, the VH CDR1 of the second antibody variable domain comprises the sequence shown in SEQ ID NO: 157. The VH CDR2 of the second antibody variable domain may further comprise the sequence of A or AI immediately preceding the sequence shown in SEQ ID NO: 159 and/or the sequence of L, LP, LPY, LPYA, LPYAD, LPYADS, LPYADSV, LPYADSVK, or LPYADSVKG immediately following the sequence shown in SEQ ID NO: 159. For example, the VH CDR2 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 158.
  • In another embodiment, the first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 407 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 405; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 406; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 439; and the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 151; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 158; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 225. The VH CDR1 of the first antibody variable domain may further comprise the sequence of A, AM or AMN immediately following the sequence shown in SEQ ID NO: 407. For example, the VH CDR1 of the first antibody variable domain may comprise the sequence shown in SEQ ID NO: 408. The VH CDR1 of the second antibody variable domain may further comprise the sequence P, PM, or PMS immediately following the sequence shown in SEQ ID NO: 151. For example, the VH CDR1 of the second antibody variable domain may comprise the sequence shown in SEQ ID NO: 157.
  • The antibodies useful in the present invention can encompass monoclonal antibodies, polyclonal antibodies, antibody fragments (e.g., Fab, Fab′, F(ab′)2, Fv, Fc, etc.), chimeric antibodies, bispecific antibodies, heteroconjugate antibodies, single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion (e.g., a domain antibody), humanized antibodies, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies. The antibodies may be murine, rat, human, or any other origin (including chimeric or humanized antibodies).
  • In some embodiments, the BCMA or CD3 antibody as described herein is a monoclonal antibody. For example, the BCMA or CD3 antibody is a humanized monoclonal antibody or a chimeric monoclonal antibody.
  • In some embodiments, the antibody comprises a modified constant region, such as, for example without limitation, a constant region that has increased potential for provoking an immune response. For example, the constant region may be modified to have increased affinity to an Fc gamma receptor such as, e.g., FcγRI, FcγRIIA, or FcγIII.
  • In some embodiments, the antibody comprises a modified constant region, such as a constant region that is immunologically inert, that is, having a reduced potential for provoking an immune response. In some embodiments, the constant region is modified as described in Eur. J. Immunol., 29:2613-2624, 1999; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 98099518. The Fc can be human IgG1, human IgG2, human IgG3, or human IgG4. The Fc can be human IgG2 containing the mutation A330P331 to S330S331 (IgG2Aa), in which the amino acid residues are numbered with reference to the wild type IgG2 sequence. Eur. J. Immunol., 29:2613-2624, 1999. In some embodiments, the antibody comprises a constant region of IgG4 comprising the following mutations (Armour et al., Molecular Immunology 40 585-593, 2003): E233F234L235 to P233V234A235 (IgG4Ac), in which the numbering is with reference to wild type IgG4. In yet another embodiment, the Fc is human IgG4 E233F234L235 to P233V234A235 with deletion G236 (IgG4Ab). In another embodiment, the Fc is any human IgG4 Fc (IgG4, IgG4Ab or IgG4Ac) containing hinge stabilizing mutation S228 to P228 (Aalberse et al., Immunology 105, 9-19, 2002). In another embodiment, the Fc can be aglycosylated Fc.
  • In some embodiments, the constant region is aglycosylated by mutating the oligosaccharide attachment residue (such as Asn297) and/or flanking residues that are part of the glycosylation recognition sequence in the constant region. In some embodiments, the constant region is aglycosylated for N-linked glycosylation enzymatically. The constant region may be aglycosylated for N-linked glycosylation enzymatically or by expression in a glycosylation deficient host cell.
  • In some embodiments, the constant region has a modified constant region that removes or reduces Fc gamma receptor binding. For example, the Fc can be human IgG2 containing the mutation D265, in which the amino acid residues are numbered with reference to the wild type IgG2 sequence (SEQ ID NO: 493). Accordingly, in some embodiments, the constant region has a modified constant region having the sequence shown in SEQ ID NO: 496:
  • ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSG
    VHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTV
    ERKCRVRCPRCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVAVSH
    EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGK
    EYKCKVSNKGLPSSIEKTISKTKGQPREPQVYTLPPSREEMTKNQVSLT
    CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSRLTVDKS
    RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK.
  • In some embodiments, the constant region has a modified constant region having the sequence shown in SEQ ID NO: 497:
  • ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSG
    VHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTV
    ERKCEVECPECPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVAVSH
    EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGK
    EYKCKVSNKGLPSSIEKTISKTKGQPREPQVYTLPPSREEMTKNQVSLT
    CEVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKS
    RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK.
  • One way of determining binding affinity of antibodies to BCMA or CD3 is by measuring binding affinity of monofunctional Fab fragments of the antibody. To obtain monofunctional Fab fragments, an antibody (for example, IgG) can be cleaved with papain or expressed recombinantly. The affinity of a BCMA Fab fragment of an antibody can be determined by surface plasmon resonance (Biacore™3000™ surface plasmon resonance (SPR) system, Biacore™, INC, Piscataway NJ) equipped with pre-immobilized streptavidin sensor chips (SA) or anti-mouse Fc or anti-human Fc using HBS-EP running buffer (0.01M HEPES, pH 7.4, 0.15 NaCl, 3 mM EDTA, 0.005% v/v Surfactant P20). Biotinylated or Fc fusion human BCMA can be diluted into HBS-EP buffer to a concentration of less than 0.5 pg/mL and injected across the individual chip channels using variable contact times, to achieve two ranges of antigen density, either 50-200 response units (RU) for detailed kinetic studies or 800-1,000 RU for screening assays. Regeneration studies have shown that 25 mM NaOH in 25% v/v ethanol effectively removes the bound Fab while keeping the activity of BCMA on the chip for over 200 injections. Typically, serial dilutions (spanning concentrations of 0.1-10× estimated KD) of purified Fab samples are injected for 1 min at 100 μL/minute and dissociation times of up to 2 hours are allowed. The concentrations of the Fab proteins are determined by ELISA and/or SDS-PAGE electrophoresis using a Fab of known concentration (as determined by amino acid analysis) as a standard. Kinetic association rates (kon) and dissociation rates (koff) are obtained simultaneously by fitting the data globally to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program. Equilibrium dissociation constant (KD) values are calculated as koff/kon. This protocol is suitable for use in determining binding affinity of an antibody to any BCMA, including human BCMA, BCMA of another mammal (such as mouse BCMA, rat BCMA, or primate BCMA), as well as different forms of BCMA (e.g., glycosylated BCMA). Binding affinity of an antibody is generally measured at 25° C., but can also be measured at 37° C.
  • The antibodies as described herein may be made by any method known in the art. For the production of hybridoma cell lines, the route and schedule of immunization of the host animal are generally in keeping with established and conventional techniques for antibody stimulation and production, as further described herein. General techniques for production of human and mouse antibodies are known in the art and/or are described herein.
  • It is contemplated that any mammalian subject including humans or antibody producing cells therefrom can be manipulated to serve as the basis for production of mammalian, including human and hybridoma cell lines. Typically, the host animal is inoculated intraperitoneally, intramuscularly, orally, subcutaneously, intraplantar, and/or intradermally with an amount of immunogen, including as described herein.
  • Hybridomas can be prepared from the lymphocytes and immortalized myeloma cells using the general somatic cell hybridization technique of Kohler, B. and Milstein, C., Nature 256:495-497, 1975 or as modified by Buck, D. W., et al., In Vitro, 18:377-381, 1982. Available myeloma lines, including but not limited to X63-Ag8.653 and those from the Salk Institute, Cell Distribution Center, San Diego, Calif., USA, may be used in the hybridization. Generally, the technique involves fusing myeloma cells and lymphoid cells using a fusogen such as polyethylene glycol, or by electrical means well known to those skilled in the art. After the fusion, the cells are separated from the fusion medium and grown in a selective growth medium, such as hypoxanthine-aminopterin-thymidine (HAT) medium, to eliminate unhybridized parent cells. Any of the media described herein, supplemented with or without serum, can be used for culturing hybridomas that secrete monoclonal antibodies. As another alternative to the cell fusion technique, EBV immortalized B cells may be used to produce the monoclonal antibodies of the subject invention. The hybridomas are expanded and subcloned, if desired, and supernatants are assayed for anti-immunogen activity by conventional immunoassay procedures (e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay).
  • Hybridomas that may be used as source of antibodies encompass all derivatives, progeny cells of the parent hybridomas that produce monoclonal antibodies specific for BCMA, CD3, or portions thereof.
  • Hybridomas that produce such antibodies may be grown in vitro or in vivo using known procedures. The monoclonal antibodies may be isolated from the culture media or body fluids, by conventional immunoglobulin purification procedures such as ammonium sulfate precipitation, gel electrophoresis, dialysis, chromatography, and ultrafiltration, if desired. Undesired activity, if present, can be removed, for example, by running the preparation over adsorbents made of the immunogen attached to a solid phase and eluting or releasing the desired antibodies off the immunogen. Immunization of a host animal with a human BCMA or CD3, or a fragment containing the target amino acid sequence conjugated to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl2, or R1N═C═NR, where R and R1 are different alkyl groups, can yield a population of antibodies (e.g., monoclonal antibodies).
  • If desired, the antibody (monoclonal or polyclonal) of interest may be sequenced and the polynucleotide sequence may then be cloned into a vector for expression or propagation. The sequence encoding the antibody of interest may be maintained in vector in a host cell and the host cell can then be expanded and frozen for future use. Production of recombinant monoclonal antibodies in cell culture can be carried out through cloning of antibody genes from B cells by means known in the art. See, e.g. Tiller et al., J. Immunol. Methods 329, 112, 2008; U.S. Pat. No. 7,314,622.
  • In an alternative, the polynucleotide sequence may be used for genetic manipulation to “humanize” the antibody or to improve the affinity, or other characteristics of the antibody. For example, the constant region may be engineered to more nearly resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. It may be desirable to genetically manipulate the antibody sequence to obtain greater affinity to BCMA or CD3 and greater efficacy in inhibiting BCMA.
  • There are four general steps to humanize a monoclonal antibody. These are: (1) determining the nucleotide and predicted amino acid sequence of the starting antibody light and heavy variable domains (2) designing the humanized antibody, i.e., deciding which antibody framework region to use during the humanizing process (3) the actual humanizing methodologies/techniques and (4) the transfection and expression of the humanized antibody. See, for example, U.S. Pat. Nos. 4,816,567; 5,807,715; 5,866,692; 6,331,415; 5,530,101; 5,693,761; 5,693,762; 5,585,089; and 6,180,370.
  • A number of “humanized” antibody molecules comprising an antigen binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent or modified rodent V regions and their associated CDRs fused to human constant regions. See, for example, Winter et al. Nature 349:293-299, 1991, Lobuglio et al. Proc. Nat. Acad. Sci. USA 86:4220-4224, 1989, Shaw et al. J Immunol. 138:4534-4538, 1987, and Brown et al. Cancer Res. 47:3577-3583, 1987. Other references describe rodent CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody constant region. See, for example, Riechmann et al. Nature 332:323-327, 1988, Verhoeyen et al. Science 239:1534-1536, 1988, and Jones et al. Nature 321:522-525, 1986. Another reference describes rodent CDRs supported by recombinantly engineered rodent framework regions. See, for example, European Patent Publication No. 0519596. These “humanized” molecules are designed to minimize unwanted immunological response toward rodent anti-human antibody molecules which limits the duration and effectiveness of therapeutic applications of those moieties in human recipients. For example, the antibody constant region can be engineered such that it is immunologically inert (e.g., does not trigger complement lysis). See, e.g. PCT Publication No. PCT/GB99/01441; UK Patent Application No. 9809951.8. Other methods of humanizing antibodies that may also be utilized are disclosed by Daugherty et al., Nucl. Acids Res. 19:2471-2476, 1991, and in U.S. Pat. Nos. 6,180,377; 6,054,297; 5,997,867; 5,866,692; 6,210,671; and 6,350,861; and in PCT Publication No. WO 01/27160.
  • The general principles related to humanized antibodies discussed above are also applicable to customizing antibodies for use, for example, in dogs, cats, primate, equines and bovines. Further, one or more aspects of humanizing an antibody described herein may be combined, e.g., CDR grafting, framework mutation and CDR mutation.
  • In one variation, fully human antibodies may be obtained by using commercially available mice that have been engineered to express specific human immunoglobulin proteins. Transgenic animals that are designed to produce a more desirable (e.g., fully human antibodies) or more robust immune response may also be used for generation of humanized or human antibodies. Examples of such technology are Xenomouse™ from Abgenix, Inc. (Fremont, CA) and HuMAb-Mouse® and TC Mouse™ from Medarex, Inc. (Princeton, NJ).
  • In an alternative, antibodies may be made recombinantly and expressed using any method known in the art. In another alternative, antibodies may be made recombinantly by phage display technology. See, for example, U.S. Pat. Nos. 5,565,332; 5,580,717; 5,733,743; and 6,265,150; and Winter et al., Annu. Rev. Immunol. 12:433-455, 1994. Alternatively, the phage display technology (McCafferty et al., Nature 348:552-553, 1990) can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors. According to this technique, antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle. Because the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. Thus, the phage mimics some of the properties of the B cell. Phage display can be performed in a variety of formats; for review see, e.g., Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3:564-571, 1993. Several sources of V-gene segments can be used for phage display. Clackson et al., Nature 352:624-628, 1991, isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice. A repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Mark et al., J. Mol. Biol. 222:581-597, 1991, or Griffith et al., EMBO J. 12:725-734, 1993. In a natural immune response, antibody genes accumulate mutations at a high rate (somatic hypermutation). Some of the changes introduced will confer higher affinity, and B cells displaying high-affinity surface immunoglobulin are preferentially replicated and differentiated during subsequent antigen challenge. This natural process can be mimicked by employing the technique known as “chain shuffling.” (Marks et al., Bio/Technol. 10:779-783, 1992). In this method, the affinity of “primary” human antibodies obtained by phage display can be improved by sequentially replacing the heavy and light chain V region genes with repertoires of naturally occurring variants (repertoires) of V domain genes obtained from unimmunized donors. This technique allows the production of antibodies and antibody fragments with affinities in the pM-nM range. A strategy for making very large phage antibody repertoires (also known as “the mother-of-all libraries”) has been described by Waterhouse et al., Nucl. Acids Res. 21:2265-2266, 1993. Gene shuffling can also be used to derive human antibodies from rodent antibodies, where the human antibody has similar affinities and specificities to the starting rodent antibody. According to this method, which is also referred to as “epitope imprinting”, the heavy or light chain V domain gene of rodent antibodies obtained by phage display technique is replaced with a repertoire of human V domain genes, creating rodent-human chimeras. Selection on antigen results in isolation of human variable regions capable of restoring a functional antigen binding site, i.e., the epitope governs (imprints) the choice of partner. When the process is repeated in order to replace the remaining rodent V domain, a human antibody is obtained (see PCT Publication No. WO 93/06213). Unlike traditional humanization of rodent antibodies by CDR grafting, this technique provides completely human antibodies, which have no framework or CDR residues of rodent origin.
  • Antibodies may be made recombinantly by first isolating the antibodies and antibody producing cells from host animals, obtaining the gene sequence, and using the gene sequence to express the antibody recombinantly in host cells (e.g., CHO cells). Another method which may be employed is to express the antibody sequence in plants (e.g., tobacco) or transgenic milk. Methods for expressing antibodies recombinantly in plants or milk have been disclosed. See, for example, Peeters, et al. Vaccine 19:2756, 2001; Lonberg, N. and D. Huszar Int. Rev. Immunol 13:65, 1995; and Pollock, et al., J Immunol Methods 231:147, 1999. Methods for making derivatives of antibodies, e.g., humanized, single chain, etc. are known in the art.
  • Immunoassays and flow cytometry sorting techniques such as fluorescence activated cell sorting (FACS) can also be employed to isolate antibodies that are specific for BCMA, CD3, or tumor antigens of interest.
  • The antibodies as described herein can be bound to many different carriers. Carriers can be active and/or inert. Examples of well-known carriers include polypropylene, polystyrene, polyethylene, dextran, nylon, amylases, glass, natural and modified celluloses, polyacrylamides, agaroses, and magnetite. The nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding antibodies, or will be able to ascertain such, using routine experimentation. In some embodiments, the carrier comprises a moiety that targets the myocardium.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies). The hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors (such as expression vectors disclosed in PCT Publication No. WO 87/04462), which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. See, e.g., PCT Publication No. WO 87/04462. The DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant regions in place of the homologous murine sequences, Morrison et al., Proc. Nat. Acad. Sci. 81:6851, 1984, or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. In that manner, “chimeric” or “hybrid” antibodies are prepared that have the binding specificity of a monoclonal antibody herein.
  • The BCMA or tumor antigen of interest antibodies as described herein can be identified or characterized using methods known in the art, whereby reduction of BCMA or other tumor antigen expression levels are detected and/or measured. In some embodiments, a BCMA antibody is identified by incubating a candidate agent with BCMA and monitoring binding and/or attendant reduction of BCMA expression levels. The binding assay may be performed with purified BCMA polypeptide(s), or with cells naturally expressing, or transfected to express, BCMA polypeptide(s). In one embodiment, the binding assay is a competitive binding assay, where the ability of a candidate antibody to compete with a known BCMA antibody for BCMA binding is evaluated. The assay may be performed in various formats, including the ELISA format.
  • Following initial identification, the activity of a candidate BCMA, CD3, or other tumor antigen antibody can be further confirmed and refined by bioassays, known to test the targeted biological activities. Alternatively, bioassays can be used to screen candidates directly. Some of the methods for identifying and characterizing antibodies are described in detail in the Examples.
  • BCMA, CD3, or other tumor antigen antibodies may be characterized using methods well known in the art. For example, one method is to identify the epitope to which it binds, or “epitope mapping.” There are many methods known in the art for mapping and characterizing the location of epitopes on proteins, including solving the crystal structure of an antibody-antigen complex, competition assays, gene fragment expression assays, and synthetic peptide-based assays, as described, for example, in Chapter 11 of Harlow and Lane, Using Antibodies, a Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1999. In an additional example, epitope mapping can be used to determine the sequence to which an antibody binds. Epitope mapping is commercially available from various sources, for example, Pepscan Systems (Edelhertweg 15, 8219 PH Lelystad, The Netherlands). The epitope can be a linear epitope, i.e., contained in a single stretch of amino acids, or a conformational epitope formed by a three-dimensional interaction of amino acids that may not necessarily be contained in a single stretch. Peptides of varying lengths (e.g., at least 4-6 amino acids long) can be isolated or synthesized (e.g., recombinantly) and used for binding assays with a BCMA, CD3, or other tumor antigen antibody. In another example, the epitope to which the BCMA, CD3, or other tumor antigen antibody binds can be determined in a systematic screening by using overlapping peptides derived from the BCMA, CD3, or other tumor antigen sequence and determining binding by the BCMA, CD3, or other tumor antigen antibody. According to the gene fragment expression assays, the open reading frame encoding BCMA, CD3, or other tumor antigen is fragmented either randomly or by specific genetic constructions and the reactivity of the expressed fragments of BCMA, CD3, or other tumor antigen with the antibody to be tested is determined. The gene fragments may, for example, be produced by PCR and then transcribed and translated into protein in vitro, in the presence of radioactive amino acids. The binding of the antibody to the radioactively labeled BCMA, CD3, or other tumor antigen fragments is then determined by immunoprecipitation and gel electrophoresis. Certain epitopes can also be identified by using large libraries of random peptide sequences displayed on the surface of phage particles (phage libraries). Alternatively, a defined library of overlapping peptide fragments can be tested for binding to the test antibody in simple binding assays. In an additional example, mutagenesis of an antigen binding domain, domain swapping experiments and alanine scanning mutagenesis can be performed to identify residues required, sufficient, and/or necessary for epitope binding. For example, domain swapping experiments can be performed using a mutant BCMA, CD3, or other tumor antigen in which various fragments of the BCMA, CD3, or other tumor antigen protein have been replaced (swapped) with sequences from BCMA from another species (e.g., mouse), or a closely related, but antigenically distinct protein (e.g., Trop-1). By assessing binding of the antibody to the mutant BCMA, CD3, or other tumor antigen, the importance of the particular BCMA, CD3, or other tumor antigen fragment to antibody binding can be assessed.
  • Yet another method which can be used to characterize a BCMA, CD3, or other tumor antigen antibody is to use competition assays with other antibodies known to bind to the same antigen, i.e., various fragments on BCMA, CD3, or other tumor antigen, to determine if the BCMA, CD3, or other tumor antigen antibody binds to the same epitope as other antibodies. Competition assays are well known to those of skill in the art.
  • An expression vector can be used to direct expression of a BCMA, CD3, or other tumor antigen antibody. One skilled in the art is familiar with administration of expression vectors to obtain expression of an exogenous protein in vivo. See, e.g., U.S. Pat. Nos. 6,436,908; 6,413,942; and 6,376,471. Administration of expression vectors includes local or systemic administration, including injection, oral administration, particle gun or catheterized administration, and topical administration. In another embodiment, the expression vector is administered directly to the sympathetic trunk or ganglion, or into a coronary artery, atrium, ventrical, or pericardium.
  • Targeted delivery of therapeutic compositions containing an expression vector, or subgenomic polynucleotides can also be used. Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al., Trends Biotechnol., 1993, 11:202; Chiou et al., Gene Therapeutics: Methods And Applications Of Direct Gene Transfer, J. A. Wolff, ed., 1994; Wu et al., J. Biol. Chem., 263:621, 1988; Wu et al., J. Biol. Chem., 269:542, 1994; Zenke et al., Proc. Natl. Acad. Sci. USA, 87:3655, 1990; and Wu et al., J. Biol. Chem., 266:338, 1991. Therapeutic compositions containing a polynucleotide are administered in a range of about 100 ng to about 200 mg of DNA for local administration in a gene therapy protocol. Concentration ranges of about 500 ng to about 50 mg, about 1 μg to about 2 mg, about 5 μg to about 500 μg, and about 20 μg to about 100 μg of DNA can also be used during a gene therapy protocol. The therapeutic polynucleotides and polypeptides can be delivered using gene delivery vehicles. The gene delivery vehicle can be of viral or non-viral origin (see generally, Jolly, Cancer Gene Therapy, 1:51, 1994; Kimura, Human Gene Therapy, 5:845, 1994; Connelly, Human Gene Therapy, 1995, 1:185; and Kaplitt, Nature Genetics, 6:148, 1994). Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence can be either constitutive or regulated.
  • Viral-based vectors for delivery of a desired polynucleotide and expression in a desired cell are well known in the art. Exemplary viral-based vehicles include, but are not limited to, recombinant retroviruses (see, e.g., PCT Publication Nos. WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; WO 93/11230; WO 93/10218; WO 91/02805; U.S. Pat. Nos. 5,219,740 and 4,777,127; GB Pat. No. 2,200,651; and EP Pat. No. 0 345 242), alphavirus-based vectors (e.g., Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249: ATCC VR-532)), and adeno-associated virus (AAV) vectors (see, e.g., PCT Publication Nos. WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655). Administration of DNA linked to killed adenovirus as described in Curiel, Hum. Gene Ther., 1992, 3:147 can also be employed.
  • Non-viral delivery vehicles and methods can also be employed, including, but not limited to, polycationic condensed DNA linked or unlinked to killed adenovirus alone (see, e.g., Curiel, Hum. Gene Ther., 3:147, 1992); ligand-linked DNA (see, e.g., Wu, J. Biol. Chem., 264:16985, 1989); eukaryotic cell delivery vehicles cells (see, e.g., U.S. Pat. No. 5,814,482; PCT Publication Nos. WO 95/07994; WO 96/17072; WO 95/30763; and WO 97/42338) and nucleic charge neutralization or fusion with cell membranes. Naked DNA can also be employed. Exemplary naked DNA introduction methods are described in PCT Publication No. WO 90/11092 and U.S. Pat. No. 5,580,859. Liposomes that can act as gene delivery vehicles are described in U.S. Pat. No. 5,422,120; PCT Publication Nos. WO 95/13796; WO 94/23697; WO 91/14445; and EP 0524968. Additional approaches are described in Philip, Mol. Cell Biol., 14:2411, 1994 and in Woffendin, Proc. Natl. Acad. Sci., 91:1581, 1994.
  • In some embodiments, the invention encompasses compositions, including pharmaceutical compositions, comprising antibodies described herein or made by the methods and having the characteristics described herein. As used herein, compositions comprise one or more antibodies that bind to CD3 and a tumor antigen (e.g. BCMA), and/or one or more polynucleotides comprising sequences encoding one or more these antibodies. These compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • The invention also provides methods of making any of these antibodies. The antibodies of this invention can be made by procedures known in the art. The polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis. Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available. For example, an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Pat. Nos. 5,807,715; 4,816,567; and 6,331,415.
  • Heteroconjugate antibodies, comprising two covalently joined antibodies, are also within the scope of the invention. Such antibodies have been used to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (PCT Publication Nos. WO 91/00360 and WO 92/200373; EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents and techniques are well known in the art, and are described in U.S. Pat. No. 4,676,980.
  • Chimeric or hybrid antibodies also may be prepared in vitro using known methods of synthetic protein chemistry, including those involving cross-linking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
  • In the recombinant humanized antibodies, the Fcγ portion can be modified to avoid interaction with Fcγ receptor and the complement and immune systems. The techniques for preparation of such antibodies are described in WO 99/58572. For example, the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. See, for example, U.S. Pat. Nos. 5,997,867 and 5,866,692.
  • The invention encompasses modifications to the antibodies and polypeptides of the invention variants as described herein, including functionally equivalent antibodies which do not significantly affect their properties and variants which have enhanced or decreased activity and/or affinity. For example, the amino acid sequence may be mutated to obtain an antibody with the desired binding affinity to BCMA and/or CD3. Modification of polypeptides is routine practice in the art and need not be described in detail herein. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or which mature (enhance) the affinity of the polypeptide for its ligand, or use of chemical analogs.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to an epitope tag. Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody of an enzyme or a polypeptide which increases the half-life of the antibody in the blood circulation.
  • Substitution variants have at least one amino acid residue in the antibody molecule removed and a different residue inserted in its place. The sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated. Conservative substitutions are shown in Table 5 under the heading of “conservative substitutions.” If such substitutions result in a change in biological activity, then more substantial changes, denominated “exemplary substitutions” in Table 5, or as further described below in reference to amino acid classes, may be introduced and the products screened.
  • TABLE 5
    Amino Acid Substitutions
    Original Residue
    (naturally occurring Conservative
    amino acid) Substitutions Exemplary Substitutions
    Ala (A) Val Val; Leu; Ile
    Arg (R) Lys Lys; Gln; Asn
    Asn (N) Gln Gln; His; Asp, Lys; Arg
    Asp (D) Glu Glu; Asn
    Cys (C) Ser Ser; Ala
    Gln (Q) Asn Asn; Glu
    Glu (E) Asp Asp; Gln
    Gly (G) Ala Ala
    His (H) Arg Asn; Gln; Lys; Arg
    Ile (I) Leu Leu; Val; Met; Ala; Phe; Norleucine
    Leu (L) Ile Norleucine; Ile; Val; Met; Ala; Phe
    Lys (K) Arg Arg; Gln; Asn
    Met (M) Leu Leu; Phe; Ile
    Phe (F) Tyr Leu; Val; Ile; Ala; Tyr
    Pro (P) Ala Ala
    Ser (S) Thr Thr
    Thr (T) Ser Ser
    Trp (W) Tyr Tyr; Phe
    Tyr (Y) Phe Trp; Phe; Thr; Ser
    Val (V) Leu Ile; Leu; Met; Phe; Ala; Norleucine
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring amino acid residues are divided into groups based on common side-chain properties:
      • (1) Non-polar: Norleucine, Met, Ala, Val, Leu, IIe;
      • (2) Polar without charge: Cys, Ser, Thr, Asn, Gln;
      • (3) Acidic (negatively charged): Asp, Glu;
      • (4) Basic (positively charged): Lys, Arg;
      • (5) Residues that influence chain orientation: Gly, Pro; and
      • (6) Aromatic: Trp, Tyr, Phe, His.
  • Non-conservative substitutions are made by exchanging a member of one of these classes for another class.
  • Any cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking. Conversely, cysteine bond(s) may be added to the antibody to improve its stability, particularly where the antibody is an antibody fragment such as an Fv fragment.
  • Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the variable region. Changes in the variable region can alter binding affinity and/or specificity. In some embodiments, no more than one to five conservative amino acid substitutions are made within a CDR domain. In other embodiments, no more than one to three conservative amino acid substitutions are made within a CDR domain. In still other embodiments, the CDR domain is CDR H3 and/or CDR L3.
  • Modifications also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation. Antibodies are glycosylated at conserved positions in their constant regions (Jefferis and Lund, Chem. Immunol. 65:111-128, 1997; Wright and Morrison, TibTECH 15:26-32, 1997). The oligosaccharide side chains of the immunoglobulins affect the protein's function (Boyd et al., Mol. Immunol. 32:1311-1318, 1996; Wittwe and Howard, Biochem. 29:4175-4180, 1990) and the intramolecular interaction between portions of the glycoprotein, which can affect the conformation and presented three-dimensional surface of the glycoprotein (Jefferis and Lund, supra; Wyss and Wagner, Current Opin. Biotech. 7:409-416, 1996). Oligosaccharides may also serve to target a given glycoprotein to certain molecules based upon specific recognition structures. Glycosylation of antibodies has also been reported to affect antibody-dependent cellular cytotoxicity (ADCC). In particular, CHO cells with tetracycline-regulated expression of P(1,4)-N-acetylglucosaminyltransferase III (GnTIII), a glycosyltransferase catalyzing formation of bisecting GIcNAc, was reported to have improved ADCC activity (Umana et al., Mature Biotech. 17:176-180, 1999).
  • Glycosylation of antibodies is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine, asparagine-X-threonine, and asparagine-X-cysteine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites). The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • The glycosylation pattern of antibodies may also be altered without altering the underlying nucleotide sequence. Glycosylation largely depends on the host cell used to express the antibody. Since the cell type used for expression of recombinant glycoproteins, e.g. antibodies, as potential therapeutics is rarely the native cell, variations in the glycosylation pattern of the antibodies can be expected (see, e.g. Hse et al., J. Biol. Chem. 272:9062-9070, 1997).
  • In addition to the choice of host cells, factors that affect glycosylation during recombinant production of antibodies include growth mode, media formulation, culture density, oxygenation, pH, purification schemes and the like. Various methods have been proposed to alter the glycosylation pattern achieved in a particular host organism including introducing or overexpressing certain enzymes involved in oligosaccharide production (U.S. Pat. Nos. 5,047,335; 5,510,261 and 5,278,299). Glycosylation, or certain types of glycosylation, can be enzymatically removed from the glycoprotein, for example, using endoglycosidase H (Endo H), N-glycosidase F, endoglycosidase F1, endoglycosidase F2, endoglycosidase F3. In addition, the recombinant host cell can be genetically engineered to be defective in processing certain types of polysaccharides. These and similar techniques are well known in the art.
  • Other methods of modification include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay. Modified polypeptides are made using established procedures in the art and can be screened using standard assays known in the art, some of which are described below and in the Examples.
  • In some embodiments of the invention, the antibody comprises a modified constant region, such as a constant region that has increased affinity to a human Fc gamma receptor, is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate macrophages; or has reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating antibody-dependent cell mediated cytotoxicity (ADCC), or activating microglia. Different modifications of the constant region may be used to achieve optimal level and/or combination of effector functions. See, for example, Morgan et al., Immunology 86:319-324, 1995; Lund et al., J. Immunology 157:4963-9 157:4963-4969, 1996; Idusogie et al., J. Immunology 164:4178-4184, 2000; Tao et al., J. Immunology 143: 2595-2601, 1989; and Jefferis et al., Immunological Reviews 163:59-76, 1998. In some embodiments, the constant region is modified as described in Eur. J. Immunol., 1999, 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 9809951.8. In other embodiments, the antibody comprises a human heavy chain IgG2 constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wild type IgG2 sequence). Eur. J. Immunol., 1999, 29:2613-2624. In still other embodiments, the constant region is aglycosylated for N-linked glycosylation. In some embodiments, the constant region is aglycosylated for N-linked glycosylation by mutating the glycosylated amino acid residue or flanking residues that are part of the N-glycosylation recognition sequence in the constant region. For example, N-glycosylation site N297 may be mutated to A, Q, K, or H. See, Tao et al., J. Immunology 143: 2595-2601, 1989; and Jefferis et al., Immunological Reviews 163:59-76, 1998. In some embodiments, the constant region is aglycosylated for N-linked glycosylation. The constant region may be aglycosylated for N-linked glycosylation enzymatically (such as removing carbohydrate by enzyme PNGase), or by expression in a glycosylation deficient host cell.
  • Other antibody modifications include antibodies that have been modified as described in PCT Publication No. WO 99/58572. These antibodies comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant region of a human immunoglobulin heavy chain. These antibodies are capable of binding the target molecule without triggering significant complement dependent lysis, or cell-mediated destruction of the target. In some embodiments, the effector domain is capable of specifically binding FcRn and/or FcγRIIb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain CH2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy.
  • The invention includes affinity matured embodiments. For example, affinity matured antibodies can be produced by procedures known in the art (Marks et al., Bio/Technology, 10:779-783, 1992; Barbas et al., Proc Nat. Acad. Sci, USA 91:3809-3813, 1994; Schier et al., Gene, 169:147-155, 1995; Yelton et al., J. Immunol., 155:1994-2004, 1995; Jackson et al., J. Immunol., 154(7):3310-9, 1995, Hawkins et al., J. Mol. Biol., 226:889-896, 1992; and PCT Publication No. WO2004/058184).
  • The following methods may be used for adjusting the affinity of an antibody and for characterizing a CDR. One way of characterizing a CDR of an antibody and/or altering (such as improving) the binding affinity of a polypeptide, such as an antibody, termed “library scanning mutagenesis”. Generally, library scanning mutagenesis works as follows. One or more amino acid positions in the CDR are replaced with two or more (such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) amino acids using art recognized methods. This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of two or more members (if two or more amino acids are substituted at every position). Generally, the library also includes a clone comprising the native (unsubstituted) amino acid. A small number of clones, e.g., about 20-80 clones (depending on the complexity of the library), from each library are screened for binding affinity to the target polypeptide (or other binding target), and candidates with increased, the same, decreased, or no binding are identified. Methods for determining binding affinity are well-known in the art. Binding affinity may be determined using Biacore™ surface plasmon resonance analysis, which detects differences in binding affinity of about 2-fold or greater. Biacore™ is particularly useful when the starting antibody already binds with a relatively high affinity, for example a KD of about 10 nM or lower. Screening using Biacore™ surface plasmon resonance is described in the Examples, herein.
  • Binding affinity may be determined using Kinexa Biocensor, scintillation proximity assays, ELISA, ORIGEN immunoassay (IGEN), fluorescence quenching, fluorescence transfer, and/or yeast display. Binding affinity may also be screened using a suitable bioassay.
  • In some embodiments, every amino acid position in a CDR is replaced (in some embodiments, one at a time) with all 20 natural amino acids using art recognized mutagenesis methods (some of which are described herein). This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of 20 members (if all 20 amino acids are substituted at every position).
  • In some embodiments, the library to be screened comprises substitutions in two or more positions, which may be in the same CDR or in two or more CDRs. Thus, the library may comprise substitutions in two or more positions in one CDR. The library may comprise substitution in two or more positions in two or more CDRs. The library may comprise substitution in 3, 4, 5, or more positions, said positions found in two, three, four, five or six CDRs. The substitution may be prepared using low redundancy codons. See, e.g., Table 2 of Balint et al., Gene 137(1):109-18, 1993.
  • The CDR may be CDRH3 and/or CDRL3. The CDR may be one or more of CDRL1, CDRL2, CDRL3, CDRH1, CDRH2, and/or CDRH3. The CDR may be a Kabat CDR, a Chothia CDR, or an extended CDR.
  • Candidates with improved binding may be sequenced, thereby identifying a CDR substitution mutant which results in improved affinity (also termed an “improved” substitution). Candidates that bind may also be sequenced, thereby identifying a CDR substitution which retains binding.
  • Multiple rounds of screening may be conducted. For example, candidates (each comprising an amino acid substitution at one or more position of one or more CDR) with improved binding are also useful for the design of a second library containing at least the original and substituted amino acid at each improved CDR position (i.e., amino acid position in the CDR at which a substitution mutant showed improved binding). Preparation, and screening or selection of this library is discussed further below.
  • Library scanning mutagenesis also provides a means for characterizing a CDR, in so far as the frequency of clones with improved binding, the same binding, decreased binding or no binding also provide information relating to the importance of each amino acid position for the stability of the antibody-antigen complex. For example, if a position of the CDR retains binding when changed to all 20 amino acids, that position is identified as a position that is unlikely to be required for antigen binding. Conversely, if a position of CDR retains binding in only a small percentage of substitutions, that position is identified as a position that is important to CDR function. Thus, the library scanning mutagenesis methods generate information regarding positions in the CDRs that can be changed to many different amino acids (including all 20 amino acids), and positions in the CDRs which cannot be changed or which can only be changed to a few amino acids.
  • Candidates with improved affinity may be combined in a second library, which includes the improved amino acid, the original amino acid at that position, and may further include additional substitutions at that position, depending on the complexity of the library that is desired, or permitted using the desired screening or selection method. In addition, if desired, adjacent amino acid position can be randomized to at least two or more amino acids. Randomization of adjacent amino acids may permit additional conformational flexibility in the mutant CDR, which may in turn, permit or facilitate the introduction of a larger number of improving mutations. The library may also comprise substitution at positions that did not show improved affinity in the first round of screening.
  • The second library is screened or selected for library members with improved and/or altered binding affinity using any method known in the art, including screening using Biacore™ surface plasmon resonance analysis, and selection using any method known in the art for selection, including phage display, yeast display, and ribosome display.
  • This invention also provides compositions comprising antibodies conjugated (for example, linked) to an agent that facilitate coupling to a solid support (such as biotin or avidin). For simplicity, reference will be made generally to antibodies with the understanding that these methods apply to any of the BCMA antibody embodiments described herein. Conjugation generally refers to linking these components as described herein. The linking (which is generally fixing these components in proximate association at least for administration) can be achieved in any number of ways. For example, a direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryl group, on one may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.
  • In another aspect, the invention provides a method of making any of the polynucleotides described herein.
  • Polynucleotides complementarity to any such sequences are also encompassed by the present invention. Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules. RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence. Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein. Variants preferably exhibit at least about 70% identity, more preferably, at least about 80% identity, yet more preferably, at least about 90% identity, and most preferably, at least about 95% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A “comparison window” as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O., 1978, A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D. G. and Sharp, P. M., 1989, CABIOS 5:151-153; Myers, E. W. and Muller W., 1988, CABIOS 4:11-17; Robinson, E. D., 1971, Comb. Theor. 11:105; Santou, N., Nes, M., 1987, Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R., 1973, Numerical Taxonomy the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, CA; Wilbur, W. J. and Lipman, D. J., 1983, Proc. Natl. Acad. Sci. USA 80:726-730.
  • Preferably, the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof. Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementarity sequence).
  • Suitable “moderately stringent conditions” include prewashing in a solution of 5×SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-65° C., 5×SSC, overnight; followed by washing twice at 65° C. for 20 minutes with each of 2×, 0.5× and 0.2×SSC containing 0.1% SDS.
  • As used herein, “highly stringent conditions” or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42° C., with washes at 42° C. in 0.2×SSC (sodium chloride/sodium citrate) and 50% formamide at 55° C., followed by a high-stringency wash consisting of 0.1×SSC containing EDTA at 55° C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
  • It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • The polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
  • For preparing polynucleotides using recombinant methods, a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein. Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome. The polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al., 1989.
  • Alternatively, PCR allows reproduction of DNA sequences. PCR technology is well known in the art and is described in U.S. Pat. Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston, 1994.
  • RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., 1989, supra, for example.
  • Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector. Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mp18, mp19, pBR322, pMB9, CoIE1, pCR1, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462. Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • The vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus). The choice of introducing vectors or polynucleotides will often depend on features of the host cell.
  • The invention also provides host cells comprising any of the polynucleotides described herein. Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest. Non-limiting examples of mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462. Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe; or K. lactis). Preferably, the host cells express the cDNAs at a level of about 5 fold higher, more preferably, 10 fold higher, even more preferably, 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells. Screening the host cells for a specific binding to BCMA or an BCMA domain (e.g., domains 1-4) is effected by an immunoassay or FACS. A cell overexpressing the antibody or protein of interest can be identified.
  • Methods of Using the Bispecific Antibodies
  • The antibodies (e.g., BCMA, CD3, or bispecific) and the antibody conjugates (e.g., BCMA antibody-drug conjugates) of the present invention are useful in various applications including, but are not limited to, therapeutic treatment methods and diagnostic treatment methods.
  • In one aspect, the invention provides a method for treating a condition associated with BCMA expression in a subject. In some embodiments, the method of treating a condition associated with BCMA expression in a subject comprises administering to the subject in need thereof an effective amount of a composition (e.g., pharmaceutical composition) comprising the BCMA antibodies or the BCMA antibody conjugates as described herein. The conditions associated with BCMA expression include, but are not limited to, abnormal BCMA expression, altered or aberrant BCMA expression, malignant cells expressing BCMA, and a proliferative disorder (e.g., cancer) or autoimmune disorder.
  • In another aspect, the invention provides a method for treating a B-cell related cancer or malignant cells expressing a tumor antigen. In some embodiments, provided is a method of treating a B-cell related cancer in a subject in need thereof comprising a) providing the bispecific antibody as described herein, and b) administering said bispecific antibody to said patient. In some embodiments, provided is a method of treating a condition associated with malignant cells expressing a tumor antigen in a subject comprising administering to the subject in need thereof an effective amount of a pharmaceutical composition comprising the bispecific antibody as described herein.
  • Accordingly, in some embodiments, provided is a method of treating a cancer in a subject comprising administering to the subject in need thereof an effective amount of a composition comprising the antibodies (e.g., BCMA, or CD3-BCMA bispecific antibodies) or the BCMA antibody conjugates as described herein. As used herein, cancer can be a B-cell related cancer including, but are not limited to, multiple myeloma, malignant plasma cell neoplasm, Hodgkin's lymphoma, nodular lymphocyte predominant Hodgkin's lymphoma, Kahler's disease and Myelomatosis, plasma cell leukemia, plasmacytoma, B-cell prolymphocytic leukemia, hairy cell leukemia, B-cell non-Hodgkin's lymphoma (NHL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), follicular lymphoma, Burkitt's lymphoma, marginal zone lymphoma, mantle cell lymphoma, large cell lymphoma, precursor B-lymphoblastic lymphoma, myeloid leukemia, Waldenstrom's macroglobulienemia, diffuse large B cell lymphoma, follicular lymphoma, marginal zone lymphoma, mucosa-associated lymphatic tissue lymphoma, small cell lymphocytic lymphoma, mantle cell lymphoma, Burkitt lymphoma, primary mediastinal (thymic) large B-cell lymphoma, lymphoplasmactyic lymphoma, Waldenström macroglobulinemia, nodal marginal zone B cell lymphoma, splenic marginal zone lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, lymphomatoid granulomatosis, T cell/histiocyte-rich large B-cell lymphoma, primary central nervous system lymphoma, primary cutaneous diffuse large B-cell lymphoma (leg type), EBV positive diffuse large B-cell lymphoma of the elderly, diffuse large B-cell lymphoma associated with inflammation, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, large B-cell lymphoma arising in HHV8-associated multicentric Castleman disease, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma, and other B-cell related lymphoma.
  • In some embodiments, provided is a method of inhibiting tumor growth or progression in a subject who has malignant cells expressing BCMA, comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein. In other embodiments, provided is a method of inhibiting metastasis cells expressing BCMA in a subject, comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein. In other embodiments, provided is a method of inducing tumor regression in malignant cells in a subject, comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein.
  • In some embodiments, provided is a method of treating an autoimmune disorder in a subject comprising administering to the subject in need thereof an effective amount of a composition comprising the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein.
  • As used herein, autoimmune disorders include, but are not limited to, systemic lupus erythematosus, rheumatoid arthritis, diabetes (Type 1), multiple sclerosis, Addison's disease, celiac disease, dermatomyositis, Graves' disease, hashimoto's thyroiditis, hashimoto's encephalopathy, Myasthenia gravis, pernicious anemia, reactive arthritis, Sjogren syndrome, acute disseminated encephalomyelitis, agammaglobulinemia, amyotrophic lateral sclerosis, ankylosing spondylitis, antiphospholipid syndrome, antisynthetase syndrome, atopic allergy, atopic dermatitis, autoimmune enteropathy, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune lymphoproliferative syndrome, autoimmune peripheral neuropathy, autoimmune pancreatitis, autoimmune polyendorcrine syndrome, autoimmune progesterone dermatitis, autoimmune thrombocytopenic purpura, autoimmune urticarial, autoimmune uveitis, Bechet's disease, Castleman's disease, cold agglutinin disease, Crohn's disease, dermatomyositis, eosinophilic fasciitis, gastrointestinal pemphigoid, Goodpasture's syndrome, Guillain-Barre syndrome, hidradenitis suppurativa, idiopathic thrombocytopenic purpura, narcolepsy, pemphigus vulgaris, pernicious anaemia, polymyositis, primary billary cirrhosis, relapsing polychrondritis, rheumatic fever, temporal arteritis, transverse myelitis, ulcerative colitis, undifferentiated connective tissue disease, vasculitis, and Wegener's granulomatosis.
  • In another aspect, the invention provides an effective amount of a composition (e.g., pharmaceutical composition) comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression in a subject in need thereof. In some embodiments, provided is an effective amount of a composition (e.g., pharmaceutical composition) comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inhibiting tumor growth or progression in a subject who has malignant cells expressing BCMA. In some embodiments, provided is an effective amount of a composition (e.g., pharmaceutical composition) comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inhibiting metastasis of malignant cells expressing BCMA in a subject in need thereof. In some embodiments, provided is an effective amount of a composition (e.g., pharmaceutical composition) comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inducing tumor regression in a subject who has malignant cells expressing BCMA.
  • In another aspect, the invention provides the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for use in treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression in a subject in need thereof. In some embodiments, provided is the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inhibiting tumor growth or progression in a subject who has malignant cells expressing BCMA. In some embodiments, provided is the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inhibiting metastasis of malignant cells expressing BCMA in a subject in need thereof. In some embodiments, provided is the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein for inducing tumor regression in a subject who has malignant cells expressing BCMA.
  • In another aspect, the invention provides a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for treating a condition (e.g., cancer or autoimmune disorder) associated with BCMA expression. In some embodiments, provided is a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting tumor growth or progression. In some embodiments, provided is a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inhibiting metastasis of malignant cells expressing BCMA. In some embodiments, provided is a use of the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein in the manufacture of a medicament for inducing tumor regression.
  • In another aspect, provided is a method of detecting, diagnosing, and/or monitoring a condition associated with BCMA expression. For example, the antibodies (e.g., BCMA or CD3-BCMA bispecific) as described herein can be labeled with a detectable moiety such as an imaging agent and an enzyme-substrate label. The antibodies as described herein can also be used for in vivo diagnostic assays, such as in vivo imaging (e.g., PET or SPECT), or a staining reagent.
  • In some embodiments, the methods described herein further comprise a step of treating a subject with an additional form of therapy. In some embodiments, the additional form of therapy is an additional anti-cancer therapy including, but not limited to, chemotherapy, radiation, surgery, hormone therapy, and/or additional immunotherapy.
  • In some embodiments, the additional form of therapy comprises administering one or more therapeutic agent in addition to the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein. The one or more therapeutic agent can be a chemotherapeutic agents including, but not limited to, a second antibody (e.g., an anti-VEGF (Vascular Endothelial Growth Factor) antibody (e.g., AVASTIN®), an anti-HER2 antibody (e.g., HERCEPTIN®), an anti-CD25 antibody, an anti-CD33 antibody, an anti-CD20 antibody (e.g., RITUXAN®), an anti-mucin-like glycoprotein antibody, an anti-TNF antibody, and/or an epidermal growth factor receptor (EGFR) antibody (e.g., ERBITUX®)), an angiogenesis inhibitor, a cytotoxic agent (e.g., anthracyclines (e.g., daunorubicin, doxorubicin, epirubicin, idarubicin, valrubicin, and mitoxantrone), taxane (e.g., paclitaxel and docetaxel), dolastatin, duocarmycin, enediyne, geldanamycin, maytansine, puromycin, vinca alkaloid (e.g., vincristine), a topoisomerase inhibitor (e.g., etoposide), tubulysin, a pyrimidine analog (e.g., fluorouracil), platinum-containing agents (e.g., cisplatin, carboplatin, and oxaliplatin), alkylating agents (e.g., melphalan, cyclophosphamide, or carmustine) and hemiasterlin), immunomodulating agent (e.g., prednisone and lenalidomide (REVLIMID®)), an anti-inflammatory agent (e.g., dexamethasone), an aromatase inhibitor (e.g., anastrozole, exemestane, letrozole, vorozole, formestane, or testolactone), a proteasome inhibitor (e.g., bortezomib such as VELCADE® ([(1R)-3-methyl-1-[[(2S)-1-oxo-3-phenyl-2-[(pyrazinylcarbonyl)amino]propy-I]amino]butyl] boronic acid or carfilzomib), and other agents such as tamoxifen.
  • For example, in some embodiments, provided is a method of treating multiple myeloma comprising administering to a patient need thereof an effective amount of a composition comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein and one or more other therapeutic agent such as a chemotherapeutic agent (e.g., doxorubicin or carfilzomib) or thalidomide or its derivative thereof (e.g., lenalidomide (REVLIMID®)). In some embodiments, the one or more other therapeutic agent is selecting from the group consisting of bortezomib (e.g., VELCADE®), melphalan, prednisone, doxorubicin, lenalidomide, thalidomide, prednisone, carmustine, etoposide, cisplatin, cyclophosphamide, carfilzomib, and vincristine. In some embodiments, the other therapeutic agent is bortezomib (e.g., VELCADE®), melphalan, lenalidomide (REVLIMID®), carfilzomib, doxorubicin, or prednisone. Accordingly, provided is a method of treating multiple myeloma comprising administering to a patient need thereof an effective amount of a composition comprising the antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein and one or more other therapeutic agent selecting from the group consisting of bortezomib, lenalidomide, carfilzomib, and doxorubicin. In some embodiments, the patient is relapsing or refractory to previous multiple myeloma therapy.
  • The antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates can be administered to an individual via any suitable route. It should be understood by persons skilled in the art that the examples described herein are not intended to be limiting but to be illustrative of the techniques available. Accordingly, in some embodiments, the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate is administered to an individual in accord with known methods, such as intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, intracranial, transdermal, subcutaneous, intra-articular, sublingually, intrasynovial, via insufflation, intrathecal, oral, inhalation or topical routes. Administration can be systemic, e.g., intravenous administration, or localized. Commercially available nebulizers for liquid formulations, including jet nebulizers and ultrasonic nebulizers are useful for administration. Liquid formulations can be directly nebulized and lyophilized powder can be nebulized after reconstitution. Alternatively, the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate can be aerosolized using a fluorocarbon formulation and a metered dose inhaler, or inhaled as a lyophilized and milled powder.
  • In one embodiment, the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate is administered via site-specific or targeted local delivery techniques. Examples of site-specific or targeted local delivery techniques include various implantable depot sources of the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate or local delivery catheters, such as infusion catheters, indwelling catheters, or needle catheters, synthetic grafts, adventitial wraps, shunts and stents or other implantable devices, site specific carriers, direct injection, or direct application. See, e.g., PCT Publication No. WO 00/53211 and U.S. Pat. No. 5,981,568.
  • Various formulations of the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate may be used for administration. In some embodiments, the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate may be administered neat. In some embodiments, the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate and a pharmaceutically acceptable excipient may be in various formulations. Pharmaceutically acceptable excipients are known in the art, and are relatively inert substances that facilitate administration of a pharmacologically effective substance. For example, an excipient can give form or consistency, or act as a diluent. Suitable excipients include but are not limited to stabilizing agents, wetting and emulsifying agents, salts for varying osmolarity, encapsulating agents, buffers, and skin penetration enhancers. Excipients as well as formulations for parenteral and nonparenteral drug delivery are set forth in Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005.
  • In some embodiments, these agents are formulated for administration by injection (e.g., intraperitoneally, intravenously, subcutaneously, intramuscularly, etc.). Accordingly, these agents can be combined with pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like. The particular dosage regimen, i.e., dose, timing and repetition, will depend on the particular individual and that individual's medical history.
  • The antibodies (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugates as described herein can be administered using any suitable method, including by injection (e.g., intraperitoneally, intravenously, subcutaneously, intramuscularly, etc.). The antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate can also be administered via inhalation, as described herein. Generally, for administration of an antibody (e.g., BCMA or CD3-BCMA bispecific) and a BCMA antibody conjugate, an initial candidate dosage can be about 2 mg/kg. For the purpose of the present invention, a typical daily dosage might range from about any of 3 pg/kg to 30 pg/kg to 300 pg/kg to 3 mg/kg, to 30 mg/kg, to 100 mg/kg or more, depending on the factors mentioned above. For example, dosage of about 1 mg/kg, about 2.5 mg/kg, about 5 mg/kg, about 10 mg/kg, and about 25 mg/kg may be used. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of symptoms occurs or until sufficient therapeutic levels are achieved, for example, to inhibit or delay tumor growth/progression or metatstasis of cancer cells. An exemplary dosing regimen comprises administering an initial dose of about 2 mg/kg, followed by a weekly maintenance dose of about 1 mg/kg of the antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate, or followed by a maintenance dose of about 1 mg/kg every other week. Other exemplary dosing regimen comprises administering increasing doses (e.g., initial dose of 1 mg/kg and gradual increase to one or more higher doses every week or longer time period). Other dosage regimens may also be useful, depending on the pattern of pharmacokinetic decay that the practitioner wishes to achieve. For example, in some embodiments, dosing from one to four times a week is contemplated. In other embodiments, dosing once a month or once every other month or every three months is contemplated. The progress of this therapy is easily monitored by conventional techniques and assays. The dosing regimen (including the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate used) can vary over time.
  • For the purpose of the present invention, the appropriate dosage of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate will depend on the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate (or compositions thereof) employed, the type and severity of symptoms to be treated, whether the agent is administered for therapeutic purposes, previous therapy, the patient's clinical history and response to the agent, the patient's clearance rate for the administered agent, and the discretion of the attending physician. Typically the clinician will administer an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate until a dosage is reached that achieves the desired result. Dose and/or frequency can vary over course of treatment. Empirical considerations, such as the half-life, generally will contribute to the determination of the dosage. For example, antibodies that are compatible with the human immune system, such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's immune system. Frequency of administration may be determined and adjusted over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of symptoms, e.g., tumor growth inhibition or delay, etc. Alternatively, sustained continuous release formulations of antibodies (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugates may be appropriate. Various formulations and devices for achieving sustained release are known in the art.
  • In one embodiment, dosages for an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate may be determined empirically in individuals who have been given one or more administration(s) of the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate. Individuals are given incremental dosages of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate. To assess efficacy, an indicator of the disease can be followed.
  • Administration of an antibody (e.g., BCMA or CD3-BCMA bispecific) or an BCMA antibody conjugate in accordance with the method in the present invention can be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners. The administration of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate may be essentially continuous over a preselected period of time or may be in a series of spaced doses.
  • In some embodiments, more than one antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate may be present. At least one, at least two, at least three, at least four, at least five different or more antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate can be present. Generally, those antibodies (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugates may have complementary activities that do not adversely affect each other. For example, one or more of the following antibody may be used: a first BCMA or CD3 antibody directed to one epitope on BCMA or CD3 and a second BCMA or CD3 antibody directed to a different epitope on BCMA or CD3.
  • Therapeutic formulations of the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate used in accordance with the present invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and may comprise buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).
  • Liposomes containing the antibody (e.g., BCMA or CD3-BCMA bispecific) or the BCMA antibody conjugate are prepared by methods known in the art, such as described in Epstein, et al., Proc. Natl. Acad. Sci. USA 82:3688, 1985; Hwang, et al., Proc. Natl Acad. Sci. USA 77:4030, 1980; and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556. Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • The active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington, The Science and Practice of Pharmacy 21st Ed. Mack Publishing, 2005.
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or ‘poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(−)-3-hydroxybutyric acid.
  • The formulations to be used for in vivo administration must be sterile. This is readily accomplished by, for example, filtration through sterile filtration membranes. Therapeutic antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate compositions are generally placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • The compositions according to the present invention may be in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, or administration by inhalation or insufflation.
  • For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention. The tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • Suitable surface-active agents include, in particular, non-ionic agents, such as polyoxyethylenesorbitans (e.g. Tween™ 20, 40, 60, 80 or 85) and other sorbitans (e.g. Span™ 20, 40, 60, 80 or 85). Compositions with a surface-active agent will conveniently comprise between 0.05 and 5% surface-active agent, and can be between 0.1 and 2.5%. It will be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
  • Suitable emulsions may be prepared using commercially available fat emulsions, such as Intralipid™, Liposyn™, Infonutrol™, Lipofundin™ and Lipiphysan™. The active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g. soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g. egg phospholipids, soybean phospholipids or soybean lecithin) and water. It will be appreciated that other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion. Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%. The fat emulsion can comprise fat droplets between 0.1 and 1.0 μm, particularly 0.1 and 0.5 μm, and have a pH in the range of 5.5 to 8.0.
  • The emulsion compositions can be those prepared by mixing an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate with Intralipid™ or the components thereof (soybean oil, egg phospholipids, glycerol and water).
  • Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulised by use of gases. Nebulised solutions may be breathed directly from the nebulising device or the nebulising device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • Compositions
  • The compositions used in the methods of the invention comprise an effective amount of an antibody (e.g., BCMA or CD3-BCMA bispecific) or a BCMA antibody conjugate as described herein. Examples of such compositions, as well as how to formulate, are also described in an earlier section and below. In some embodiments, the composition comprises one or more antibodies (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugates. For example, BCMA antibody or CD3-BCMA bispecific antibody recognizes human BCMA or CD3-BCMA. In some embodiments, the BCMA or CD3-BCMA antibody is a human antibody, a humanized antibody, or a chimeric antibody. In some embodiments, the BCMA antibody or CD3-BCMA antibody comprises a constant region that is capable of triggering a desired immune response, such as antibody-mediated lysis or ADCC. In other embodiments, the BCMA antibody or CD3-BCMA antibody comprises a constant region that does not trigger an unwanted or undesirable immune response, such as antibody-mediated lysis or ADCC.
  • It is understood that the compositions can comprise more than one antibody (e.g., BCMA or CD3-BCMA bispecific) or BCMA antibody conjugate (e.g., a mixture of BCMA antibodies or CD3-BCMA bispecific antibodies that recognize different epitopes of BCMA or CD3 and BCMA). Other exemplary compositions comprise more than one BCMA antibody, CD3-BCMA antibody, or BCMA antibody conjugate that recognize the same epitope(s), or different species of BCMA antibodies, CD3-BCMA bispecific antibodies, or BCMA antibody conjugate that bind to different epitopes of BCMA (e.g., human BCMA) or CD3 and BCMA (human CD3 and BCMA).
  • The composition used in the present invention can further comprise pharmaceutically acceptable carriers, excipients, or stabilizers (Remington: The Science and practice of Pharmacy 21st Ed., 2005, Lippincott Williams and Wilkins, Ed. K. E. Hoover), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). Pharmaceutically acceptable excipients are further described herein.
  • Kits
  • The invention also provides kits for use in the instant methods. Kits of the invention include one or more containers comprising the BCMA antibody, CD3-BCMA bispecific antibody, or the BCMA antibody conjugate as described herein and instructions for use in accordance with any of the methods of the invention described herein. Generally, these instructions comprise a description of administration of the BCMA antibody, CD3-BCMA bispecific antibody, or the BCMA antibody conjugate for the above described therapeutic treatments.
  • The instructions relating to the use of the BCMA antibodies, CD3-BCMA bispecific antibodies, or the BCMA antibody conjugates as described herein generally include information as to dosage, dosing schedule, and route of administration for the intended treatment. The containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
  • The kits of this invention are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Also contemplated are packages for use in combination with a specific device, such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump. A kit may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The container may also have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a BCMA antibody, CD3-BCMA bispecific antibody, or a BCMA antibody conjugate. The container may further comprise a second pharmaceutically active agent.
  • Kits may optionally provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container.
  • The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims.
  • Examples Example 1: Determination of Kinetics and Affinity of hBCMA/Human IgG Interactions at 25° C. and/or 37° C.
  • This example determines the kinetics and affinity of various anti-BCMA antibodies at 25° C. and 37° C.
  • All experiments were performed on a Bio-Rad Proteon XPR36 surface Plasmon resonance biosensor (Bio-Rad, Hercules, CA). An array of anti-BCMA antibodies was prepared using an amine-coupling method on a Bio-Rad GLC Sensor Chip similar to that described in Abdiche, et al., Anal. Biochem. 411, 139-151 (2011). The analysis temperature for the immobilization was 25° C. and the running buffer was HBS-T+(10 mM HEPES, 150 mM NaCl, 0.05% Tween-20, pH 7.4). Channels were activated in the analyte (horizontal) direction by injecting a mixture of 1 mM ECD and 0.25 mM NHS for 3 minutes at a flow rate of 30 μL/min. IgGs were immobilized on the activated spots by injecting them in the ligand (vertical) direction at 20 pg/mL in 10 mM Acetate pH 4.5 buffer for 1.5 minutes at 30 pg/mL. The activated surfaces were blocked by injecting 1M ethanolamine, pH 8.5 in the analyte direction for 3 minutes at 30 pL/min.
  • The analysis temperature for the hBCMA binding analysis was 37° C. or 25° C. in a running buffer of HBS-T+, supplemented with 1 mg/mL BSA. A kinetic titration method was employed for the interaction analysis as described in Abdiche, et al. The hBCMA (human BCMA) analyte was injected in the analyte direction using a series of injections from low to high concentration. The concentrations used were 0.08 nM, 0.4 nM, 2 nM, 10 nM and 50 nM (a 5-membered series, with a 5-fold dilution factor and top concentration of 50 nM). The association time for a given analyte dilution was two minutes. Immediately after the 50 nM hBCMA injection, dissociation was monitored for 2 hours. Prior to the hBCMA analyte injections, buffer was injected 5 times using the same association and dissociation times at the hBCMA analyte cycles to prepare a buffer blank sensorgram for double-referencing purposes (double referencing as described in Myszka, J. Mol. Recognit. 12, 279-284 (1999).
  • The sensorgrams were double-referenced and fit to a 1:1 Langmuir with mass transport kinetic titration model in BIAevaluation Software version 4.1.1 (GE Lifesciences, Piscataway, NJ). The sensorgrams and fits are shown in FIG. 1 , and the kinetics and affinity parameters for various anti-BCMA antibodies of the present invention are shown in Tables 6A-6C.
  • TABLE 6A
    ka kd t1/2 KD
    Sample (1/Ms) (1/s) (min) (pM)
    A02_Rd4_6 nM_C01 1.2E+06 2.8E−05 411 24
    A02_Rd4_6 nM_C16 1.1E+06 6.2E−05 187 59
    Combo_Rd4_0.6 nM_C29 6.6E+06 1.4E−04 83 21
    L3PY/H3TAQ 2.6E+06 1.4E−04 84 53
  • TABLE 6B
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    huBCMA huBCMA huBCMA huBCMA
    Antibody @ 25° C. @25° C. @25° C. @ 25° C.
    P6E01/P6E01 1.04E+06 4.15E−03 2.8 4.0
    P6E01/H3.AQ 8.35E+05 3.45E−04 33.53 0.41
    L1.LGF/L3.KW/P6E01 8.31E+05 7.55E−03 1.53 9.08
    L1.LGF/L3.NY/P6E01 1.33E+06 4.40E−03 2.63 3.32
    L1.GDF/L3.NY/P6E01 1.60E+06 5.92E−03 1.95 3.70
    L1.LGF/L3.KW/H3.AL 4.28E+05 1.23E−03 9.40 2.87
    L1.LGF/L3.KW/H3.AP 9.28E+05 2.27E−03 5.10 2.44
    L1.LGF/L3.KW/H3.AQ 5.24E+05 9.56E−04 12.09 1.82
    L1.LGF/L3.PY/H3.AP 4.57E+05 9.69E−04 11.92 2.12
    L1.LGF/L3.PY/H3.AQ 9.31E+05 8.86E−04 13.04 0.95
    L1.LGF/L3.NY/H3.AL 7.63E+05 9.70E−04 11.91 1.27
    L1.LGF/L3.NY/H3.AP 9.36E+05 5.33E−04 21.67 0.57
    L1.LGF/L3.NY/H3.AQ 6.66E+05 2.99E−04 38.61 0.45
    L1.GDF/L3.KW/H3.AL 4.45E+05 3.90E−03 2.96 8.76
    L1.GDF/L3.KW/H3.AP 1.17E+06 4.61E−03 2.51 3.93
    L1.GDF/L3.KW/H3.AQ 7.97E+05 3.48E−03 3.32 4.37
    L1.GDF/L3.PY/H3.AQ 1.42E+06 1.35E−02 0.86 9.49
    L1.GDF/L3.NY/H3.AL 9.07E+05 4.03E−03 2.87 4.44
    L1.GDF/L3.NY/H3.AP 1.41E+06 1.41E−03 8.21 1.00
    L1.GDF/L3.NY/H3.AQ 9.84E+05 7.22E−04 16.00 0.73
    L3.KW/P6E01 7.40E+05 3.15E−04 36.66 0.43
    L3.PY/P6E01 7.12E+05 2.28E−04 50.74 0.32
    L3.NY/P6E01 8.76E+05 3.84E−04 30.08 0.44
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    huBCMA huBCMA huBCMA huBCMA
    Antibody @ 37° C. @37° C. @37° C. @ 37° C.
    L3.PY/L1.PS/P6E01 2.49E+06 1.13E−03 10.21 0.45
    L3.PY/L1.AH/P6E01 2.55E+06 1.26E−03 9.19 0.49
    L3.PY/L1.FF/P6E01 2.39E+06 1.41E−03 8.18 0.59
    L3.PY/L1.PH/P6E01 2.81E+06 9.13E−04 12.65 0.32
    L3.PY/L3.KY/P6E01 3.18E+06 1.09E−03 10.65 0.34
    L3.PY/L3.KF/P6E01 2.88E+06 2.08E−03 5.56 0.72
    L3.PY/H2.QR 2.56E+06 1.19E−03 9.75 0.46
    L3.PY/H2.DY 2.60E+06 1.38E−03 8.37 0.53
    L3.PY/H2.YQ 2.58E+06 1.56E−03 7.41 0.60
    L3.PY/H2.LT 2.40E+06 1.29E−03 8.95 0.54
    L3.PY/H2.HA 2.43E+06 1.47E−03 7.89 0.60
    L3.PY/H2.QL 2.64E+06 2.18E−03 5.31 0.82
    L3.PY/H3.YA 3.15E+06 1.18E−03 9.82 0.37
    L3.PY/H3.AE 3.29E+06 1.39E−03 8.32 0.42
    L3.PY/H3.AQ 3.08E+06 1.73E−03 6.69 0.56
    L3.PY/H3.TAQ 3.08E+06 1.14E−03 10.13 0.37
    L3.PY/P6E01 2.65E+06 1.96E−03 5.91 0.74
    L3.PY/L1.PS/H2.QR 3.97E+06 1.03E−01 0.11 25.85
    L3.PY/L1.PS/H2.DY 3.22E+06 3.61E−03 3.20 1.12
    L3.PY/L1.PS/H2.YQ 3.35E+06 4.30E−03 2.69 1.28
    L3.PY/L1.PS/H2.LT 3.40E+06 4.65E−03 2.49 1.37
    L3.PY/L1.PS/H2.HA 3.30E+06 1.06E−02 1.09 3.21
    L3.PY/L1.PS/H2.QL 1.52E+07 3.14E−01 0.04 20.64
    L3.PY/L1.PS/H3.YA 3.07E+06 9.05E−03 1.28 2.95
    L3.PY/L1.PS/H3.AE 3.14E+06 1.46E−03 7.93 0.46
    L3.PY/L1.PS/H3.AQ 3.26E+06 1.79E−03 6.46 0.55
    L3.PY/L1.PS/H3.TAQ 3.25E+06 2.46E−03 4.70 0.76
    L3.PY/L1.AH/H2.QR 3.13E+06 1.81E−03 6.39 0.58
    L3.PY/L1.AH/H2.DY 3.05E+06 1.52E−03 7.62 0.50
    L3.PY/L1.AH/H2.YQ 2.42E+06 1.93E−03 6.00 0.80
    L3.PY/L1.AH/H2.LT 3.16E+06 1.23E−03 9.38 0.39
    L3.PY/L1.AH/H2.HA 3.33E+06 1.81E−03 6.37 0.54
    L3.PY/L1.AH/H2.QL 3.04E+06 1.60E−03 7.22 0.53
    L3.PY/L1.AH/H3.YA 3.00E+06 1.50E−03 7.73 0.50
    L3.PY/L1.AH/H3.AE 3.32E+06 1.73E−03 6.70 0.52
    L3.PY/L1.AH/H3.AQ 3.03E+06 1.97E−03 5.85 0.65
    L3.PY/L1.AH/H3.TAQ 3.27E+06 1.19E−03 9.68 0.37
    L3.PY/L1.FF/H2.QR 3.47E+06 1.77E−03 6.54 0.51
    L3.PY/L1.FF/H2.DY 4.14E+06 2.71E−03 4.27 0.65
    L3.PY/L1.FF/H2.YQ 3.32E+06 1.52E−03 7.61 0.46
    L3.PY/L1.FF/H2.LT 3.30E+06 1.67E−03 6.92 0.51
    L3.PY/L1.FF/H2.HA 3.49E+06 2.19E−03 5.29 0.63
    L3.PY/L1.FF/H2.QL 3.48E+06 1.40E−03 8.28 0.40
    L3.PY/L1.FF/H3.YA 3.50E+06 1.80E−03 6.41 0.51
    L3.PY/L1.FF/H3.AE 3.82E+06 2.63E−03 4.39 0.69
    L3.PY/L1.FF/H3.AQ 3.32E+06 1.54E−03 7.51 0.46
    L3.PY/L1.FF/H3.TAQ 3.52E+06 1.89E−03 6.12 0.54
    L3.PY/L1.PH/H2.QR 3.69E+06 2.36E−03 4.89 0.64
    L3.PY/L1.PH/H2.HA 2.37E+06 1.16E−03 9.99 0.49
    L3.PY/L1.PH/H3.AE 3.68E+06 1.34E−03 8.61 0.36
    L3.PY/L1.PH/H3.AQ 3.08E+06 1.59E−03 7.27 0.52
    L3.PY/L1.PH/H3.TAQ 3.58E+06 2.13E−03 5.43 0.59
    L3.PY/L3.KY/H2.QR 2.95E+06 9.90E−04 11.67 0.34
    L3.PY/L3.KY/H2.DY 3.19E+06 6.42E−04 18.00 0.20
    L3.PY/L3.KY/H2.YQ 2.14E+06 1.65E−03 7.02 0.77
    L3.PY/L3.KY/H2.LT 2.92E+06 9.06E−04 12.75 0.31
    L3.PY/L3.KY/H2.HA 3.29E+06 1.63E−03 7.10 0.49
    L3.PY/L3.KY/H2.QL 3.65E+06 2.08E−03 5.56 0.57
    L3.PY/L3.KY/H3.YA 3.30E+06 9.12E−04 12.67 0.28
    L3.PY/L3.KY/H3.TAQ 2.79E+06 6.49E−04 17.79 0.23
    L3.PY/L3.KF/H2.DY 2.74E+06 1.82E−03 6.35 0.67
    L3.PY/L3.KF/H2.YQ 1.96E+06 2.23E−03 5.18 1.14
    L3.PY/L3.KF/H2.LT 2.75E+06 1.91E−03 6.05 0.69
    L3.PY/L3.KF/H2.QL 2.07E+06 1.25E−03 9.26 0.60
    L3.PY/L3.KF/H3.YA 3.12E+06 1.47E−03 7.85 0.47
    L3.PY/L3.KF/H3.AE 3.07E+06 1.55E−03 7.44 0.51
    L3.PY/L3.KF/H3.AQ 3.48E+06 2.27E−03 5.09 0.65
    L3.PY/L3.KF/H3.TAQ 2.82E+06 1.62E−03 7.12 0.58
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    cyBCMA cyBCMA cyBCMA cyBCMA
    Antibody @ 25° C. @25° C. @25° C. @ 25° C.
    P6E01/P6E01 7.02E−02 0.16 115.4
    P6E01/H3.AQ 1.08E+06 7.40E−03 1.6 6.9
    L1.LGF/L3.KW/P6E01 4.55E+05 1.95E−02 0.6 42.8
    L1.LGF/L3.NY/P6E01 9.20E+05 1.05E−02 1.1 11.4
    L1.GDF/L3.NY/P6E01 1.20E+06 7.67E−03 1.5 6.4
    L1.LGF/L3.KW/H3.AL 2.90E+05 1.21E−02 1.0 41.8
    L1.LGF/L3.KW/H3.AP 5.54E+05 1.54E−02 0.7 27.8
    L1.LGF/L3.KW/H3.AQ 5.27E+05 3.55E−03 3.3 6.7
    L1.LGF/L3.PY/H3.AP 3.64E+05 1.30E−02 0.9 35.8
    L1.LGF/L3.PY/H3.AQ 1.00E+06 4.77E−03 2.4 4.8
    L1.LGF/L3.NY/H3.AL 6.35E+05 1.48E−02 0.8 23.2
    L1.LGF/L3.NY/H3.AP 8.30E+05 5.57E−03 2.1 6.7
    L1.LGF/L3.NY/H3.AQ 7.51E+05 1.48E−03 7.8 2.0
    L1.GDF/L3.KW/H3.AL 3.18E+05 1.80E−02 0.6 56.7
    L1.GDF/L3.KW/H3.AP 8.14E+05 2.03E−02 0.6 24.9
    L1.GDF/L3.KW/H3.AQ 8.02E+05 5.65E−03 2.0 7.0
    L1.GDF/L3.PY/H3.AQ 1.55E+06 1.66E−02 0.7 10.7
    L1.GDF/L3.NY/H3.AL 9.00E+05 2.19E−02 0.5 24.3
    L1.GDF/L3.NY/H3.AP 1.36E+06 7.02E−03 1.6 5.2
    L1.GDF/L3.NY/H3.AQ 1.18E+06 1.36E−03 8.5 1.2
    L3.KW/P6E01 7.63E+05 2.57E−03 4.5 3.4
    L3.PY/P6E01 8.55E+05 2.93E−03 3.9 3.4
    L3.NY/P6E01 1.01E+06 2.87E−03 4.0 2.8
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    cyBCMA cyBCMA cyBCMA cyBCMA
    Antibody @ 37° C. @37° C. @37° C. @ 37° C.
    L3.PY/L1.PS/P6E01 2.17E+06 6.06E−03 1.91 2.79
    L3.PY/L1.AH/P6E01 2.16E+06 5.72E−03 2.02 2.65
    L3.PY/L1.FF/P6E01 2.45E+06 5.91E−03 1.96 2.41
    L3.PY/L1.PH/P6E01 2.17E+06 7.89E−03 1.46 3.63
    L3.PY/L3.KY/P6E01 2.27E+06 5.02E−03 2.30 2.21
    L3.PY/L3.KF/P6E01 2.39E+06 8.30E−03 1.39 3.48
    L3.PY/H2.QR 2.18E+06 6.58E−03 1.76 3.02
    L3.PY/H2.DY 2.24E+06 6.18E−03 1.87 2.76
    L3.PY/H2.YQ 2.46E+06 6.21E−03 1.86 2.53
    L3.PY/H2.LT 2.09E+06 7.57E−03 1.53 3.63
    L3.PY/H2.HA 1.99E+06 7.55E−03 1.53 3.79
    L3.PY/H2.QL 2.05E+06 1.26E−02 0.91 6.16
    L3.PY/H3.YA 2.87E+06 5.40E−03 2.14 1.88
    L3.PY/H3.AE 2.82E+06 5.04E−03 2.29 1.79
    L3.PY/H3.AQ 2.77E+06 5.39E−03 2.14 1.94
    L3.PY/H3.TAQ 2.57E+06 4.37E−03 2.64 1.70
    L3.PY/P6E01 2.20E+06 1.31E−02 0.88 5.96
    L3.PY/L1.PS/H2.QR 5.25E+05 6.70E−04 17.23 1.28
    L3.PY/L1.PS/H2.DY 1.90E+06 3.78E−03 3.06 1.99
    L3.PY/L1.PS/H2.YQ 2.00E+06 3.74E−03 3.09 1.87
    L3.PY/L1.PS/H2.LT 2.17E+06 4.11E−03 2.81 1.89
    L3.PY/L1.PS/H2.HA 1.45E+06 2.69E−03 4.30 1.86
    L3.PY/L1.PS/H2.QL 6.57E+05 6.36E−04 18.17 0.97
    L3.PY/L1.PS/H3.YA 1.77E+06 9.98E−03 1.16 5.65
    L3.PY/L1.PS/H3.AE 2.46E+06 4.13E−03 2.80 1.68
    L3.PY/L1.PS/H3.AQ 2.52E+06 4.33E−03 2.67 1.72
    L3.PY/L1.PS/H3.TAQ 2.58E+06 5.52E−03 2.09 2.14
    L3.PY/L1.AH/H2.QR 2.20E+06 4.91E−03 2.35 2.23
    L3.PY/L1.AH/H2.DY 2.32E+06 4.51E−03 2.56 1.95
    L3.PY/L1.AH/H2.YQ 1.58E+06 4.31E−03 2.68 2.74
    L3.PY/L1.AH/H2.LT 2.19E+06 2.96E−03 3.91 1.35
    L3.PY/L1.AH/H2.HA 2.58E+06 4.39E−03 2.63 1.70
    L3.PY/L1.AH/H2.QL 2.62E+06 9.55E−03 1.21 3.65
    L3.PY/L1.AH/H3.YA 2.37E+06 5.26E−03 2.20 2.22
    L3.PY/L1.AH/H3.AE 2.25E+06 3.56E−03 3.25 1.58
    L3.PY/L1.AH/H3.AQ 2.24E+06 3.99E−03 2.90 1.78
    L3.PY/L1.AH/H3.TAQ 2.28E+06 3.02E−03 3.83 1.32
    L3.PY/L1.FF/H2.QR 2.55E+06 4.21E−03 2.75 1.65
    L3.PY/L1.FF/H2.DY 2.66E+06 5.00E−03 2.31 1.88
    L3.PY/L1.FF/H2.YQ 2.19E+06 3.26E−03 3.55 1.49
    L3.PY/L1.FF/H2.LT 2.19E+06 3.41E−03 3.38 1.56
    L3.PY/L1.FF/H2.HA 2.33E+06 4.17E−03 2.77 1.79
    L3.PY/L1.FF/H2.QL 2.36E+06 4.49E−03 2.57 1.91
    L3.PY/L1.FF/H3.YA 2.46E+06 4.16E−03 2.77 1.69
    L3.PY/L1.FF/H3.AE 2.85E+06 5.01E−03 2.31 1.76
    L3.PY/L1.FF/H3.AQ 2.18E+06 3.29E−03 3.51 1.51
    L3.PY/L1.FF/H3.TAQ 2.32E+06 3.76E−03 3.07 1.62
    L3.PY/L1.PH/H2.QR 2.42E+06 4.36E−03 2.65 1.80
    L3.PY/L1.PH/H2.HA 1.61E+06 5.53E−03 2.09 3.44
    L3.PY/L1.PH/H3.AE 2.61E+06 2.02E−03 5.72 0.77
    L3.PY/L1.PH/H3.AQ 2.28E+06 3.41E−03 3.39 1.50
    L3.PY/L1.PH/H3.TAQ 2.51E+06 3.20E−03 3.61 1.28
    L3.PY/L3.KY/H2.QR 2.05E+06 7.74E−03 1.49 3.78
    L3.PY/L3.KY/H2.DY 1.96E+06 2.43E−03 4.75 1.24
    L3.PY/L3.KY/H2.YQ 1.27E+06 2.58E−03 4.47 2.04
    L3.PY/L3.KY/H2.LT 1.82E+06 2.32E−03 4.98 1.27
    L3.PY/L3.KY/H2.HA 2.28E+06 3.18E−03 3.63 1.40
    L3.PY/L3.KY/H2.QL 2.75E+06 4.09E−03 2.83 1.49
    L3.PY/L3.KY/H3.YA 1.84E+06 4.28E−03 2.70 2.33
    L3.PY/L3.KY/H3.TAQ 1.81E+06 1.92E−03 6.03 1.06
    L3.PY/L3.KF/H2.DY 2.08E+06 3.68E−03 3.14 1.77
    L3.PY/L3.KF/H2.YQ 1.41E+06 5.01E−03 2.30 3.55
    L3.PY/L3.KF/H2.LT 1.91E+06 4.13E−03 2.80 2.16
    L3.PY/L3.KF/H2.QL 1.42E+06 3.10E−03 3.73 2.18
    L3.PY/L3.KF/H3.YA 2.10E+06 7.96E−03 1.45 3.78
    L3.PY/L3.KF/H3.AE 1.85E+06 5.64E−03 2.05 3.05
    L3.PY/L3.KF/H3.AQ 2.55E+06 2.38E−03 4.85 0.93
    L3.PY/L3.KF/H3.TAQ 2.01E+06 1.91E−03 6.05 0.95
  • TABLE 6C*
    Human Cyno
    KD KD
    Antibody ka (1/Ms) kd (1/s) (pM) ka (1/Ms) kd (1/s) (pM)
    P5A2_VHVL 6.96E+06 3.87E−02 5567 1.61E+06 1.64E−02 10230
    A02_Rd4_0.6nM_C06 3.49E+06 7.37E−05 21 1.81E+06 1.05E−04 58
    A02_Rd4_0.6nM_C09 5.50E+06 9.75E−05 18 2.13E+06 1.74E−04 82
    A02_Rd4_6nM_C16 1.56E+06 1.41E−04 90 1.34E+06 1.58E−04 118
    A02_Rd4_6nM_C03 1.69E+06 1.26E−04 75 1.17E+06 1.85E−04 158
    A02_Rd4_6nM_C01 3.11E+06 9.20E−05 30 1.45E+06 5.83E−04 401
    A02_Rd4_6nM_C26 4.26E+06 1.39E−04 33 2.21E+06 4.48E−04 203
    A02_Rd4_6nM_C25 2.75E+06 1.80E−04 65 1.50E+06 3.30E−04 220
    A02_Rd4_6nM_C22 3.38E+06 1.82E−04 54 1.84E+06 3.24E−04 176
    A02_Rd4_6nM_C19 3.00E+06 1.48E−04 49 2.54E+06 6.61E−04 260
    A02_Rd4_0.6nM_C03 4.27E+06 1.82E−04 43 2.12E+06 4.26E−04 201
    A02_Rd4_6nM_C07 1.48E+06 1.89E−04 128 6.91E+05 7.86E−04 1138
    A02_Rd4_6nM_C23 1.22E+07 2.55E−04 21 2.63E+06 4.14E−04 157
    A02_Rd4_0.6nM_C18 4.73E+06 2.29E−04 48 3.24E+06 6.39E−04 197
    A02_Rd4_6nM_C10 4.51E+06 3.15E−04 70 1.90E+06 8.98E−04 472
    A02_Rd4_6nM_C05 3.10E+06 3.08E−04 99 1.36E+06 1.29E−03 950
    A02_Rd4_0.6nM_C10 2.30E+06 2.96E−04 129 8.83E+05 1.63E−03 1842
    A02_Rd4_6nM_C04 4.47E+06 6.03E−04 135 2.18E+06 8.31E−04 381
    A02_Rd4_0.6nM_C26 7.26E+06 4.43E−04 61 2.71E+06 2.56E−03 941
    A02_Rd4_0.6nM_C13 8.53E+06 5.66E−04 66 2.29E+06 1.28E−03 560
    A02_Rd4_0.6nM_C01 4.74E+06 9.15E−04 193 2.39E+06 1.57E−03 655
    A02_Rd4_6nM_C08 3.92E+06 7.38E−04 188 2.23E+06 1.13E−02 5072
    P5C1_VHVL 1.16E+07 6.92E−02 5986 3.53E+06 5.38E−02 15231
    C01_Rd4_6nM_C24 7.47E+06 3.48E−03 467 3.17E+06 8.91E−04 281
    C01_Rd4_6nM_C26 1.50E+07 1.36E−03 90 4.75E+06 1.99E−03 419
    C01_Rd4_6nM_C02 1.61E+07 1.44E−03 89 5.12E+06 2.18E−03 426
    C01_Rd4_6nM_C10 1.31E+07 2.12E−03 162 4.44E+06 2.19E−03 493
    C01_Rd4_0.6nM_C27 1.23E+07 3.74E−03 303 3.34E+06 2.85E−03 852
    C01_Rd4_6nM_C20 6.02E+06 2.76E−03 459 3.60E+06 6.25E−03 1737
    C01_Rd4_6nM_C12 1.21E+07 6.49E−03 535 4.51E+06 3.70E−03 820
    C01_Rd4_0.6nM_C16 1.55E+07 6.30E−03 407 4.95E+06 4.64E−03 939
    C01_Rd4_0.6nM_C09 1.51E+07 8.25E−03 545 5.28E+06 9.36E−03 1773
    C01_Rd4_6nM_C09 1.58E+07 1.28E−02 811 3.73E+06 8.68E−03 2328
    C01_Rd4_0.6nM_C03 1.55E+07 1.50E−02 964 4.72E+06 1.19E−02 2528
    C01_Rd4_0.6nM_C06 1.82E+07 1.54E−02 847 6.22E+06 1.21E−02 1948
    C01_Rd4_6nM_C04 2.33E+07 4.97E−02 2134 6.34E+06 3.27E−02 5156
    COMBO_Rd4_0.6nM_C22 1.97E+06 7.15E−05 36 1.34E+06 6.66E−05 50
    COMBO_Rd4_6nM_C21 1.17E+07 7.34E−05 6 3.17E+06 2.48E−04 78
    COMBO_Rd4_6nM_C10 5.47E+06 9.72E−05 18 1.52E+06 1.60E−04 105
    COMBO_Rd4_0.6nM_C04 1.07E+07 1.58E−04 15 3.52E+06 1.37E−04 39
    COMBO_Rd4_6nM_C25 7.98E+06 1.13E−04 14 2.85E+06 2.26E−04 79
    COMBO_Rd4_0.6nM_C21 1.34E+07 1.15E−04 9 3.63E+06 3.04E−04 84
    COMBO_Rd4_6nM_C11 6.74E+06 1.24E−04 18 2.64E+06 4.12E−04 156
    COMBO_Rd4_0.6nM_C20 7.65E+06 1.46E−04 19 3.09E+06 2.84E−04 92
    COMBO_Rd4_6nM_C09 8.85E+06 1.43E−04 16 2.37E+06 3.18E−04 134
    COMBO_Rd4_6nM_C08 8.99E+06 1.69E−04 19 3.06E+06 4.28E−04 140
    COMBO_Rd4_0.6nM_C19 7.86E+06 1.55E−04 20 2.92E+06 9.79E−04 336
    COMBO_Rd4_0.6nM_C02 8.57E+06 1.85E−04 22 3.01E+06 4.94E−04 164
    COMBO_Rd4_0.6nM_C23 7.39E+06 2.10E−04 28 2.81E+06 5.31E−04 189
    COMBO_Rd4_0.6nM_C29 1.47E+07 2.77E−04 19 4.00E+06 3.36E−04 84
    COMBO_Rd4_0.6nM_C09 1.04E+07 3.19E−04 31 3.77E+06 3.46E−04 92
    COMBO_Rd4_6nM_C12 1.38E+07 2.70E−04 20 3.29E+06 4.86E−04 148
    COMBO_Rd4_0.6nM_C30 4.35E+06 2.82E−04 65 1.68E+06 8.08E−04 481
    COMBO_Rd4_0.6nM_C14 8.66E+06 3.28E−04 38 3.48E+06 6.45E−04 185
    COMBO_Rd4_6nM_C07 1.05E+07 3.71E−04 35 3.94E+06 9.34E−04 237
    COMBO_Rd4_6nM_C02 1.05E+06 4.43E−04 422 7.95E+05 1.36E−03 1714
    COMBO_Rd4_0.6nM_C05 4.32E+06 4.97E−04 115 1.94E+06 1.72E−03 886
    COMBO_Rd4_0.6nM_C17 8.68E+06 8.01E−04 92 3.06E+06 1.01E−03 330
    COMBO_Rd4_6nM_C22 3.03E+06 7.75E−04 256 1.70E+06 1.65E−03 972
    COMBO_Rd4_0.6nM_C11 5.11E+06 1.06E−03 207 2.20E+06 4.23E−03 1924
    *The binding analysis was conducted at 37° C.
  • Example 2: Flow Cytometry of Human Anti-BCMA Antibodies on BCMA Positive Tumor Cells
  • This example demonstrates binding of BCMA positive tumor cells by various BCMA antibodies of the present invention.
  • Binding of human anti-hBCMA expressed in mouse IgG2a were assessed on BCMA-expressing cells (KMS12BM, L363, MM1S and KMS12PE) by flow cytometry. 250,000 cells were incubated with 0.5 ug antibody in 100 uL binding buffer (PBS (Phosphate Buffered Saline)+0.2% BSA (Bovine Serum Albumin)), followed by incubation with Alex Fluor 647 conjugated anti-mouse IgG (Biolegend). Table 7 shows MFI (mean fluorescence intensity) on BCMA positive tumor cells by various BCMA antibodies (e.g., Combo_Rd4_0.6 nM_C29, A02_Rd4_6 nM_C01, A02_Rd4_6 nM_C16, and P6E01/H3TAQ)
  • TABLE 7
    Combo_Rd4_0.6 A02_Rd4_6 A02_Rd4_6 P6E01/
    secondary only nM_C29 nM_C01 nM H3TAQ
    Cell Line MFI
    KMS12PE (BCMA+++) 26 6114 5862 3094 6018
    MM1S (BCMA++) 22 2569 2539 1951 2715
    L363 (BCMA+) 22 1667 1176 789 1457
    KMS12BM (BCMA+) 22 583 580 421 634
  • Example 3: Cytotoxicity of Anti-BCMA ADCs in BCMA Positive Cells
  • This example illustrates the efficacy of the anti-BCMA ADCs in BCMA positive cells.
  • Human anti-BCMA (L3.PY/P6E01, L3.PY/H3.TAQ, Combo_Rd4_0.6 nM_C29, A02_Rd4_6 nM_C01, and A02_Rd4_6 nM C16) antibodies were expressed as human IgG1 subtypes engineered with glutamine-containing transglutaminase (“Q”) tags (e.g. LCQ05, H7c, N297A, N297Q, N297A/H7c, N297Q/LCQ05) for drug antibody ratios (DAR) of 2, 4, and 6. TG17 corresponds to SEQ ID NO: 472 (LLQGPP); LCQ05 correspond to SEQ ID NO: 474 (GGLLQGPP), H7c correspond to SEQ ID NO: 454 (LLQG), respectively, and conjugated with AcLys-Val-Cit-PABC-Aur0101 (Acetyl-Lysine-Valine-Citrulline-p-aminobenzyloxycarbonyl), amino-PEG6-C2-Aur3377, or amino-PEG6-C2-Aur0131 as indicated in Table 8. In one instance, the transglutaminase tags can be engineered at the light chain, heavy chain, or a combination of light and heavy chains. In other instance, the transglutaminase tag (e.g., Q) is engineered at site of the antibody, such as at position 297 of the human IgG (EU numbering scheme). For example, the wild-type amino acid asparagine (N) is substituted with glutamine or alanine at position 297 of the BCMA antibody (N297Q or N297A) of the present invention. Anti-BCMA antibody conjugation to Aur0101, Aur3377, and Aur0131 was then achieved via microbial transglutaminase-catalyzed transamidation reaction between the anti-BCMA antibody carrying a targeted glutamine or glutamine tag at the specific site (e.g., carboxyl terminus or amino terminus of the heavy chain or light chain, position 297, or at another site of the antibody) and an amine-containing derivative of the payload (e.g., MMAD, Aur0101, Aur3377, or Aur0131). In some instances, the wild-type amino acid lysine at position 222, 340, or 370 (in accordance with EU numbering scheme) was replaced with amino acid arginine (“K222R”, “K340R”, or “K370R”). For example, the K222R substitution was found to have the surprising effect of resulting in more homogenous antibody and payload conjugate, better intermolecular crosslinking between the antibody and the payload, and/or significant decrease in interchain crosslinking with the glutamine tag on the C-terminus of the antibody light chain.
  • In the transamidation reaction, the glutamine on the antibody acted as an acyl donor, and the amine-containing compound acted as an acyl acceptor (amine donor). Purified anti-BCMA antibody in the concentration of 1-150 μM was incubated with a 5-100 molar excess acyl acceptor, ranging between 5 μM-15 mM, in the presence of 0.23-0.55% (w/v) Streptoverticillium mobaraense transglutaminase (ACTIVA™, Ajinomoto, Japan) in 10-1000 mM NaCl, and 25 mM MES, HEPES [4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid] or Tris HCl buffer at pH range 6.2-8.8. The reaction conditions were adjusted for individual acyl acceptor derivatives, and the optimal efficiency and specificity were typically observed for 33 μM antibody, 0.67 mM derivative, and 0.378% (w/v) transglutaminase in 75 mM NaCl, 25 mM Tris HCl, pH 8.5. Following incubation at 20-37 deg C. for 1-24 hours, the antibody was purified on Butyl Sepharose High Performance (Butyl HP) resin (GE Healthcare, Waukesha, WI) using standard chromatography methods known to persons skilled in the art, such as commercial hydrophobic interaction chromatography from GE Healthcare.
  • Target expressing (MM1.S, KMS12BM and L363) cells were then seeded on clear bottom plates at 3000 cells/well. Cells were treated with 4-fold serially diluted antibody-drug conjugates in triplicates. Cell viability was determined by CellTiter-Glo® Luminescent Cell Viability Assay 96 (Promega, Madison WI) 96 hours after treatment. Relative cell viability was determined as percentage of untreated control. EC50 was calculated by Prism software. Table 8 shows that all human anti-BCMA antibodies of the present invention conjugated to cytotoxic agent 0101, 3377, and 0131 through transglutaminase tags and linkers exert potent cell killing activity in BCMA expressing cells.
  • TABLE 8
    MM1.S LUCGFP KMS12BM L363
    EC50 (BCMA++) LUCGFP (+) LUCGFP (+)
    ANTIBODY DAR nM ug/mL nM ug/mL nM ug/mL
    L3.PY/P6E01 4 0.42 0.06 31.79 4.77 7.34 1.10
    N297Q/K222R AcLys
    vc0101
    L3PY/H3.TAQ 4 0.12 0.02 3.94 0.59 0.60 0.09
    N297Q/K222R Aclys
    vc0101
    A02_Rd4_6nM_C01 3.91 0.26 0.04 18.67 2.80 2.53 0.38
    N297Q/K222R Aclys
    vc0101
    A02_Rd4_6nM_C16 3.92 0.80 0.12 38.73 5.81 9.68 1.45
    N297Q/K222R Aclys
    vc0101
    Combo_Rd4_0.6nM_C29
    2 0.13 0.02 10.91 1.64 1.67 0.25
    LCQ05/K222R Aclys
    vc0101
    Combo_Rd4_0.6nM_C29 3.9 0.11 0.02 0.99 0.15 0.66 0.10
    N297Q/K222R Aclys
    vc0101
    Combo_Rd4_0.6nM_C29 5.98 0.34 0.05 1.19 0.18 1.05 0.16
    LCQ05/N297Q/K222R
    Aclys vc0101
    Combo_Rd4_0.6nM_C29 3.81 0.23 0.03 0.85 0.13 0.81 0.12
    H7c/N297A/K222R
    AmPEG6 Aur0131
    Combo_Rd4_0.6nM_C29 3.88 0.30 0.05 5.01 0.75 1.36 0.20
    H7c/N297A/K222R
    AmPEG6 Aur3377
  • Example 4: Anti-BCMA ADCs Induce Tumor Regression in an Orthotopic Multiple Myeloma Model
  • This example illustrates the in vivo efficacy of the anti-BCMA ADCs in the MM1 S orthotopic multiple myeloma model.
  • In vivo efficacy study of BCMA ADCs was performed with multiple myeloma cell line MM1.S expressing luciferase and GFP (Green Fluorescent Protein) in an orthotopic model. Ten million MM1.S LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female CB17/SCID animals. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enable monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 1-3E6 for all animals, the animals were randomized into groups and a single dose of a human anti-BCMA antibody conjugated with 1) LCQ05/K222R-vc0101 at the C-terminus of the antibody light chain and control conjugates were administered through bolus tail vein injection. Animals were terminated when they exhibit hindlimb paralysis, an endpoint for MM1.S orthotopic models. FIG. 2 shows that a single dose at 3 mg/kg of various human anti-BCMA ADCs inhibits tumor progression as compared to the negative control (NNC), including P6E01/P6E01-AcLys-Val-Cit-PABC-Aur0101, P5A2_VHVL-AcLys-Val-Cit-PABC-Aur0101; P5C1 VHVL-AcLys-Val-Cit-PABC-Aur0101; P4G4-AcLys-Val-Cit-PABC-Aur0101; and P1A11-AcLys-Val-Cit-PABC-Aur0101.
  • This study demonstrates that treatment with a BCMA-ADC inhibits progression of multiple myeloma.
  • Example 5: Anti-BCMA ADCs Induce Tumor Regression and Inhibition in an Orthotopic Multiple Myeloma Model
  • This example also illustrates the in vivo efficacy of the anti-BCMA ADCs in the MM1.S orthotopic multiple myeloma models.
  • In vivo efficacy study of BCMA ADCs was performed with multiple myeloma cell line MM1.S expressing luciferase and GFP in an orthotopic model. Ten million MM1.S LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female CB17/SCID animals. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enable monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 1-3E6 for all animals, the animals were randomized into groups; 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-Aur0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101, and 6) control conjugate LCQ04/K222R-AcLys-Val-Cit-PABC-Aur0101. A single dose of human anti-BCMA ADCs and control conjugate were administered through bolus tail vein injection. Animals were terminated when they exhibit hindlimb paralysis, an endpoint for MM1.S orthotopic model. FIG. 3 shows that a single dose of human anti-BCMA L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-0131 and 2) H7c/N297A/K222R-amino-PEG6-C2-3377 resulted in tumor regression. A single dose of human anti-BCMA L3.PY/P6E01 antibody conjugated with 1) N297Q/K222R-AcLys-Val-Cit-PABC-Aur0101, 2) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, and 3) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101 resulted in tumor inhibition.
  • Accordingly, this study demonstrates that treatment with a BCMA-ADC induces regression and inhibits progression of multiple myeloma.
  • Example 6: Anti-BCMA ADCs Induce Tumor Inhibition in an Orthotopic Multiple Myeloma Model
  • This example also illustrates the in vivo efficacy of the anti-BCMA ADCs in the KMS12BM orthotopic multiple myeloma models
  • In vivo efficacy study of BCMA ADCs was performed with multiple myeloma cell line KMS12BM expressing luciferase and GFP in an orthotopic model. 6-8 weeks old female NSG animals were irradiated with 100cGy and 24 hours post irradiation, ten million KMS12BM LucGFP cells were injected intravenously through the tail vein. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enable monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin are captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 5E6 for all animals, the animals were randomized into groups; 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-Aur010, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101, and 6) control conjugate LCQ04/K222R-AcLys-Val-Cit-PABC-Aur0101. A single dose of human anti-BCMA ADCs and control conjugate was administered through bolus tail vein injection. Animals were terminated when they lose more than 15% of total body weight, an endpoint for KMS12BM orthotopic models. FIG. 4 shows that a single dose of human anti-BCMA L3.PY/P6E01 antibody conjugated with 1) H7c/N297A/K222R-amino-PEG6-C2-3377, 2) N297Q/K222R-AcLys-Val-Cit-PABC-Aur0101, 3) LCQ05/K222R-AcLys-Val-Cit-PABC-Aur0101, 4) H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) N297Q/K222R/LCQ05-AcLys-Val-Cit-PABC-Aur0101 resulted in tumor inhibition.
  • Accordingly, this study further demonstrates that treatment with a BCMA-ADC induces regression and inhibits progression of multiple myeloma.
  • Example 7: Dose Response Curve of Anti-BCMA ADC in MM1 S Orthotopic Model
  • This example further illustrates the in vivo efficacy of the anti-BCMA ADCs in the MM1 S orthotopic multiple myeloma models
  • In vivo efficacy study of BCMA ADCs was performed with multiple myeloma cell line MM1.S expressing luciferase and GFP in an orthotopic model. Ten million MM1.S LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female CB17/SCID animals. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enable monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 1.2E6 for all animals, the animals were randomized into groups; 1) 0.1 mg/kg H7c/N297A/K222R-amino-PEG6-C2-0131, 2) 0.38 mg/kg H7c/N297A/K222R-amino-PEG6-C2-0131, 3) 0.75 mg/kg H7c/N297A/K222-amino-PEG6-C2-0131, 4) 1.5 mg/kg H7c/N297A/K222R-amino-PEG6-C2-0131, and 5) 3 mg/kg control conjugate N297Q/K222R-AcLys-VC-0101. A single dose of human anti-BCMA ADCs and control conjugate were administered through bolus tail vein injection. Animals were terminated when they exhibit hindlimb paralysis, an endpoint for MM1.S orthotopic model. FIG. 5 shows that a single dose of human anti-BCMA COMBO_Rd4_0.6 nM_C29 antibody conjugated with groups 1)-4) above resulted in tumor regression starting at 0.1 mg/kg and tumor inhibition up to 100 days starting at 0.75 mg/kg.
  • Accordingly, this study demonstrates that treatment with a BCMA-ADC induces tumor regression and tumor inhibition in multiple myeloma.
  • Example 8: Generation and Purification of Heterodimeric Antibodies
  • This example describes the generation and purification of the heterodimeric antibodies of the present application.
  • The variable region of the human specific anti-CD3 antibody was cloned into a human IgG1 or IgG2AA containing the following mutations 221R, 228R, and K409R; or 223R, 225R, 228R, and K409R, respectively, and referred as hIgG1 RRR or IgG2AA-RRRR.
  • The variable region of the anti-target antibody was cloned into a human IgG1 or IgG2AA containing the following mutations 221E, 228E, L368E or 223E, 225E, 228E and L368E, respectively, and referred as hIgG1EEE or hIgG2AA-EEEE.
  • Heterodimers were prepared by incubation of the anti-CD3 IgG1 or IgG2AA having hIgG1 RRR or IgG2AA-RRRR mutations with an anti-target antibody having hIgG1 EEE or hIgG2AA-EEEE mutations in PBS with 1 mM or 2 mM GSH for 24 hrs at 37° C. as described in International Patent Application No. PCT/US2011/036419 (WO2011/143545). The heterodimer was purified by ion exchange chromatography, as described below.
  • All the heterodimers were purified by ion exchange chromatography. Briefly, analytical ion exchange separation of the Fc-hetero and Fc-homodimers was carried out on Agilent 1100 quaternary pump LC system (Agilent Inc, Santa Clara, CA, USA) equipped with weak cation exchange DIONEX Propac WCX-10G (4×50 mm) column. Proteins were injected in 5% buffer A (20 mM MES pH 5.4) and eluted in a gradient from 25% to 75% buffer B (20 mM MES pH 5.4 and 500 mM NaCl) over a 20 minute period with 1 ml/min flow rate. Larger scale Fc-heterodimer purification was performed on an Akta Explorer (GE) equipped with weak cation exchange DIONEX Propac WCX-10G (4×250 mm) column. Proteins were injected in 5% buffer A (20 mM MES pH 5.4) and eluted in a gradient from 15% to 75% buffer B (20 mM MES pH 5.4 and 500 mM NaCl) over a 60 minute period with 1 ml/min flow rate.
  • Example 9: Determination of Kinetics and Affinity of hCD3/Human IgG Interactions at 25° C. and/or 37° C.
  • This example determines the kinetics and affinity of various anti-CD3 antibodies at 25° C. and 37° C.
  • All experiments were performed on a Bio-Rad Proteon XPR36 surface Plasmon resonance biosensor (Bio-Rad, Hercules, CA). An array of anti-CD3 antibodies was prepared using an amine-coupling method on a Bio-Rad GLC Sensor Chip similar to that described in Abdiche, et al., Anal. Biochem. 411, 139-151 (2011). The analysis temperature for the immobilization was 25° C. and the running buffer was HBS-T+(10 mM HEPES, 150 mM NaCl, 0.05% Tween-20, pH 7.4). Channels were activated in the analyte (horizontal) direction by injecting a mixture of 1 mM ECD and 0.25 mM NHS for 3 minutes at a flow rate of 30 μL/min. IgGs were immobilized on the activated spots by injecting them in the ligand (vertical) direction at 20 pg/mL in 10 mM Acetate pH 4.5 buffer for 1.5 minutes at 30 μg/mL. The activated surfaces were blocked by injecting 1M ethanolamine, pH 8.5 in the analyte direction for 3 minutes at 30 μL/min.
  • The analysis temperature for the hCD3 binding analysis was 37° C. or 25° C. in a running buffer of HBS-T+, supplemented with 1 mg/mL BSA. A kinetic titration method was employed for the interaction analysis as described in Abdiche, et al. The hCD3 (human CD3) analyte was injected in the analyte direction using a series of injections from low to high concentration. The concentrations used were 0.08 nM, 0.4 nM, 2 nM, 10 nM and 50 nM (a 5-membered series, with a 5-fold dilution factor and top concentration of 50 nM). The association time for a given analyte dilution was two minutes. Immediately after the 50 nM hCD3 injection, dissociation was monitored for 2 hours. Prior to the hCD3 analyte injections, buffer was injected 5 times using the same association and dissociation times at the hCD3 analyte cycles to prepare a buffer blank sensorgram for double-referencing purposes (double referencing as described in Myszka, J. Mol. Recognit. 12, 279-284 (1999).
  • The sensorgrams were double-referenced and fit to a 1:1 Langmuir with mass transport kinetic titration model in BIAevaluation Software version 4.1.1 (GE Lifesciences, Piscataway, NJ). The kinetics and affinity parameters for various anti-CD3 antibodies of the present invention are shown in Table 9.
  • TABLE 9
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    huCD3ed huCD3ed huCD3ed huCD3ed
    Antibody @25° C. @25° C. @25° C. @25° C.
    H2B4 3.7E+05 2.0E−03 5.8 5.3
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    huCD3ed huCD3ed huCD3ed huCD3ed
    Antibody @37° C. @37° C. @37° C. @37° C.
    H2B4 4.37E+05 0.01369 0.84 3.14E−08
    h2B4-VH-wt VL_TK 3.80E+05 1.40E−02 0.83 3.80E−08
    h2B4-VH-Hnps VL_TK 3.90E+05 1.60E−02 0.72 4.10E−08
    h2B4-VH-yads VL_TK 2.40E+05 2.10E−02 0.79 8.60E−08
    h2B4-VH-yaes VL_TK 2.30E+05 2.30E−02 0.50 1.00E−07
    h2B4-VH-yaps VL_TK 2.50E+05 2.30E−02 0.50 9.20E−08
    T½ (min) KD (nM)
    ka (1/Ms) kd (1/s) to to
    cyCD3ed cyCD3ed cyCD3ed cyCD3ed
    Antibody @ 25° C. @25° C. @25° C. @ 25° C.
    H2B4 3.9E+05 1.5E−03 7.7 3.8
  • Example 10: Flow Cytometry of Human Anti-CD3 Bispecific Antibodies on B Cells and CD8+ T Cells
  • This example demonstrates the efficacy of the anti-CD3-anti-CD20 bispecific antibodies in CD20+ cells.
  • Cynomolgus monkey studies were conducted at Charles River Laboratories, Preclinical Services Nevada in accordance with the Institutional Animal Care and Use Committee. Animals (n=2) were dosed via intravenous bolus injection with the bispecific anti-CD20/h2B4 antibody at doses of 500 ug/kg, 100 ug/kg, 20 ug/kg, 2 ug/kg, 0.2 ug/kg or 0.02 ug/kg. Animals were observed twice daily and at each blood collection time point. Blood for flow cytometry and cytokine analysis was collected into K2EDTA tubes from a peripheral vessel not used for i.v. dosing.
  • Efficacy was determined by measuring B cells and T cells in peripheral blood by flow cytometry. Whole blood was collected at the time points indicated and kept at 4° C. until analysis. Erythrocytes were lysed with ACK buffer (Gibco) for 5 minutes at room temperature and white blood cells were pelleted by centrifugation. Cells were stained for 1 hr. at 4° C. with a cocktail containing fluorescently labeled antibodies recognizing cyno CD19 (Beckman Coulter), CD45, CD4, CD8, Ki67 (BD Biosciences) in PBS+2% FBS. For Ki67 analysis, cells were first stained with CD4 and CD8, then fix/permeabilized with BD cyotfix/cytoperm kit (BD Biosciences) according to manufacturer's instructions prior to intracellular staining for Ki67. Acquisition of cells on a BD LSRII flow cytometer was carried out immediately after staining.
  • The resulting B cell count was graphed as a percentage of the pre-study B cell count in FIGS. 6A-6F. Prolonged B cell depletion following a single dose was achieved with doses as low as 2 ug/kg. B cell depletion was seen at all doses. The duration of the depletion effect was dose dependent.
  • The resulting CD8+ T cell count was graphed as percentage of the pre-study CD8+ T cell count in FIGS. 7A-7F. After an initial relocalization, T cell levels were restored to baseline levels or above for the duration of the study.
  • Example 11: Flow Cytometry of Human Anti-CD3 Bispecific Antibodies on CD8+ T Cells
  • This example demonstrates the efficacy of the monovalent anti-CD3 antibody on T cell kinetics and activation.
  • Cynomolgus monkey studies were conducted and efficacy was determined by measuring T cells in peripheral blood by flow cytometry as described in Example 3. Cynomolgus monkeys (n=2) were dosed weekly, i.v., at 0.2 ug/kg with anti-CD20/h2B4 or NNC (non-specific antibody)/h2B4. In contrast to the CD20 targeted bispecific antibody, the NNC/h2B4 has little to no effect on CD8+ T cell kinetics in the blood as measured by flow cytometry. Ki67 was used as a marker for T cell activation.
  • The resulting T cell count was graphed as percentage of the pre-study CD8+ T cell count in FIGS. 8A and 8B. In cynomolgus monkeys dosed with the CD20/h2B4 bispecific antibody, Ki67+ T cells increased and peaked between day 3 and day 7 post dose, indicating T cell activation. However, in cynomolgus monkeys dosed with NNC/h2B4, there was no increase in Ki67+ T cells.
  • Example 12: Flow Cytometry of Human Anti-CD3 Bispecific Antibodies on B Cells
  • This example demonstrates the effect of anti-CD3 arm affinity on B cell depletion.
  • Cynomolgus monkey studies were conducted and efficacy was determined by measuring T cells in peripheral blood by flow cytometry as described in Example 10. Bispecific antibodies were made with an anti-CD20 arm paired with 4 anti-CD3 antibody arms with different affinities. Following a single, i.v. dose at 0.2 ug/kg, efficacy was determined by measuring B cells in peripheral blood by flow cytometry.
  • In FIGS. 9A-9D, the resulting B cell count was graphed as a percentage of the pre-study B cell count. Efficacy of B cell depletion correlates to anti-CD3 arm affinity.
  • Example 13: In Vitro Study of the Bispecific Antibody on T-Cell Mediated Killing of BCMA Positive Cells
  • This example illustrates the in vitro cytotoxicity of the Anti-BCMA/CD3 hIgG2AA Bispecific in BCMA Positive Cells.
  • Human anti-BCMA (P5A2, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C16, P5C1, C01_Rd4_6 nM_C12, COMBO_Rd4_0.6 nM_C22, Combo_Rd4_0.6 nM_C29, L3PY/H3TAQ and A02_Rd4_6 nM_C01) and human anti-CD3 (H2B4) antibodies were expressed as human IgG2dA engineered with EEEE for bispecific exchange as described in Example 8.
  • CD3+ T cells from PBMC were negatively selected using Pan T Cell Isolation kit, human (Miltenyi, San Diego CA). Target expressing (KMS12PE, L363 and Molp8) cells and CD3+ T-cells were seeded on clear U-bottom plates at 20000 and 100000 cells/well respectively. Cells were treated with 10-fold serially diluted bispecific antibody in triplicates. Cell death was determined by CytoTox 96© Non-Radioactive Cytotoxicity Assay (Promega, Madison WI) 20 hours after treatment. Cell cytotoxicity was determined as percentage of untreated effector plus target control wells. EC50 was calculated by Prism software. Table 10 shows that all human anti-BCMA_H2B4 bispecific antibodies exert cell killing activity in BCMA expressing cells.
  • TABLE 10
    KMS12PE KMS12BM MOLP8
    Anti-BCMA bispecific (BCMA+++) (BCMA+) (MOLP8+)
    P5A2 0.371 1.509 5.231
    A02_Rd4_0.6 nM_C01 0.073 0.078 0.550
    A02_Rd4_6 nM_C16 0.186 0.052 0.315
    P5C1 0.581 4.117 N/A
    C01_Rd4_6 nM_C12 0.189 0.415 0.850
    COMBO_Rd4_0.6 nM_C22 0.115 0.049 0.065
    Combo_Rd4_0.6 nM_C29 0.175 0.049 0.201
    L3PY/H3TAQ 0.070 0.055 0.337
    A02_Rd4_6 nM_C01 UND 0.057 0.060
    UND is undetermined; N/A is EC50 could not be determined.
  • Example 14: In Vitro Characterization of the Mouse Hybridoma Cloned Anti-CD3 Antibody
  • This example illustrates the in vitro T cell activation/proliferation of the anti-CD3 cloned from mouse hybridoma in human/cynomolgus PBMC cells for antibody screening.
  • Human anti-CD3 antibodies were cloned from immunized mouse, expressed as mouse IgG1, and purified by Protein A affinity beads. Human/Cynomolgus peripheral blood mononuclear cells (hu/cyPBMC) were prepared by Ficoll (Density: 1.083 g/mL, GE) density gradient centrifugation from blood filters obtained from local blood banks. Erythrocytes were removed by incubating in LCK buffer (155 mM NH4Cl, 10 mM KHCO3, 100 mM EDTA; Gibco) for 3 minutes at room temperature. Cells were centrifuged for 5 min at 600 g. The supernatant containing the lysed erythrocytes was discarded, and the PBMC were washed twice in 50 ml 1×PBS/1% BSA/1 mM EDTA. The pelleted cells were adjusted to 107 cells per ml in culture media, X-VIVO-15, serum free media (Lonza,), and the PBMC were seeded as 106 (100ul) per-well to round bottom 96 well tissue culture plates. Selected Abs are 10× serial diluted from 1000ng to 1ng per mL for mixing with human PBMC and 5× serial diluted from 5000ng to 200ng per mL for mixing with cynomolgus PBMC. For analysis of PBMC T cell proliferation by 3H-thymidine incorporation, 2 day cultures were performed in triplicate. During the final 16 h of culture 3H-thymidine (0.5 mCi/well) was added, and incorporation was measured. Cells are harvested and lysed, DNA is captured onto glass-fiber filter. Radioactivity (cpm) as measure for proliferation by counting on a scintillation beta-counter.
  • FIGS. 10A and 10B show that the selected anti-CD3 1A4, 1C10, 2B4, and 7A3 antibodies had Thymidine incorporation reading on human and cynomolgus PBMC (peripheral blood mononuclear cells). Table 11 shows their KDs by Biacore measurement. In vitro characterization shows anti-CD3 1C10 and 2B4 antibodies are similar to the positive control SP34 anti-CD3 antibody (BD Biosciences)
  • TABLE 11
    anti-xCD3e ab/bsc_hCD3ed kinetic results
    for data fitted from 80 nM-0.64 nM
    Ligand ka kd t1/2 (min) KD (nM)
    UCHT1 (+) 1.80E+05 <8.55E−04  >13.5 <4.74
    2B4 3.74E+05 2.74E−03 4.21 7.33
    1C10 2.96E+05 2.37E−03 4.88 8.00
    SP34 (+) 2.84E+05 3.04E−03 3.80 10.73
    7A3* 82.70
    1A4* 99.97
    Note:
    Data is only reported for satisfactory kinetic fits.
    (+) = positive controls
    *Kinetic determinations are rough estimates because antibody is heterogeneous. Only steady state affinity is measured
  • Example 15: In Vitro Study of the Bispecific Antibody on T Cell Mediated Killing
  • This example illustrates the in vitro cytotoxicity of the anti-EpCam/CD3 Bispecific in SW480 mixed with healthy donor isolated Pan T cells.
  • A: Anti-CD antibodies h2B4-1d, TK, hnpsTK, and yaesTK
  • Human anti-CD3 (h2B4-1d (or h2B4), h2B4-TK (or h2B4-VH-wt VL_TK), h2B4-hnpsTK (or h2B4-VH-hnps VL_TK), and h2B4-yaesTK (or h2B4-VH-yaes VL_TK)) antibodies and human anti-EpCam antibodies were expressed as human IgG2dA engineered with RRRR or EEEE for bispecific exchange as describe in Example 8.
  • The SW480 was selected as target cell line for cell killing assay and the effector cells and human T cells were purified from human peripheral blood mononuclear cells (huPBMC). Target and effector cells were seeded in 96-well, round bottom plates in cell culture medium containing 5% fetal bovine serum (FBS). The number of target cells was kept constant at 2×104 cells/well. A 10-fold serial dilution of bispecific antibody, from 3ug to 3 μg per mL, was added in triplicate to the cells. Total reaction volume was 200 uL. The reactions were incubated for 48 and 72 hours. For the analysis of cytotoxicity, the lactate dehydrogenase (LDH), a stable cytosolic enzyme that was released upon cell lysis, was quantitatively measured by CytoTox 96® Non-Radioactive Cytotoxicity Assay kit (Promega, G1780). The plate was read on a Vmax kinetic microplate reader (Molecular Devices) at 490 nM. Optical density values were corrected for media background and spontaneous lysis of target and effector cells. Specific cytotoxicity was calculated according to the following formula:

  • [% spec. lysis=490 nM readout of sample−of E+T mixed control)/(490 nM readout of total T lysis−of media control)×100%]
  • FIGS. 11A and 11B show that all human anti-EpCam_h2B4 bispecific antibodies had cell killing activity on in vitro setting, and antibody mediated T cell activation was monitoring by T cell activation marker. Table 12A shows their EC50. Table 12B shows Biacore KD on the bispecific antibody format.
  • TABLE 12A
    Day 2: EC50 (nM)
    h2B4-1d_Ep 537.1
    h2B4-TK_Ep 405.3
    h2B4-hnpsTK_Ep 424.7
    h2B4-yaesTK_Ep 1126
  • TABLE 12B
    Summary Table for anti-CD3 hIgG2dA
    bispecifics kinetics at 37° C.
    Sample ID - bschIgG2dA ka (1/Ms) kd (1/s) t½ (min) KD (nM)
    h2B4-1d_Ep 5.86E+05 2.37E−02 0.49 40.4
    h2B4-TK_Ep 6.87E+05 2.13E−02 0.54 31.0
    h2B4-hnpsTK_Ep 7.54E+05 2.35E−02 0.49 31.2
    h2B4-yaesTK_Ep 4.74E+05 2.58E−02 0.45 54.4

    B: Anti-CD antibodies m25A8, h25A8-B12, and h25A8-B13
  • Human anti-CD3 h2B4(h2B4_1d) and h25A8 (m25A8, h25A8-B12, and h25A8-B13) and human anti-EpCam antibodies were expressed as human IgG2dA engineered with RRRR or EEEE for bispecific exchange as describe in Example 8.
  • FIGS. 11C and 11D show that all human anti-EpCam_anti-CD3 bispecific antibodies had cell killing activity on in vitro setting, and antibody mediated T cell activation was monitoring by T cell activation marker. Table 12C shows the EC50 of in vitro cell killing. Table 12D shows the Biacore kinetics on the bispecific antibody format at 37° C. Table 12E shows in vitro characterization using SEC-MALS (Size Exclusion Chromatography with Multi-Angle Light Scattering) and DSC (Differential Scanning Calorimety).
  • TABLE 12C
    EC50 of in vitro cell killing
    Day 1: EC50 (nM)
    h2B4-1d_Ep 52.9
    m25A8_Ep 127.6
    h25A8-B12_Ep 99.36
    h25A8-B13_Ep 57.11
  • TABLE 12D
    Summary Table for anti-CD3 hIgG2dA
    bispecifics kinetics at 37° C.
    SampleID -bschIgG2dA ka (1/Ms) kd (1/s) t½ (min) KD (nM)
    h25A8-B5_Ep 1.29E+06 5.81E−02 0.20 45.0
    h25A8-B8_Ep 1.19E+06 2.22E−02 0.52 18.7
    h25A8-B12_Ep 1.15E+06 2.41E−02 0.48 21.0
    h25A8-B13_Ep 1.20E+06 2.19E−02 0.53 18.3
    h25A8-C8_Ep 1.20E+06 3.01E−02 0.38 25.1
    h2B4-1d_Ep 5.86E+05 2.37E−02 0.49 40.4
  • TABLE 12E
    in vitro characterization
    DSC
    SEC-MALS Tm1 Tm2 Tm3
    % % C. C. C.
    Monomer Aggregate (CH2) (Fab) (CH3)
    m28A8 99.9 0.1 66.3 67.8 76.1
    hIgG2dA_h25A8-B12 99.2 0.8 70.91 73.06 78.31
    hIgG2dA_h25A8-B13 99.5 0.5 70.88 72.86 78.2
    hIgG2dA-h2B4TK 98.8 1.4 70.2 74.5 79.4
  • This example illustrates the in vitro cytotoxicity of the anti-BCMA/CD3 Bispecific in primary myeloma cells.
  • Human anti-BCMA (P5A2, A02_Rd4_0.6 nM_C01, A02_Rd4_6 nM_C16, Combo_Rd4_0.6 nM_029, and P6E01 L3PY/H3TAQ) and human anti-CD3 (h2B4) antibodies were expressed as human IgG2dA engineered with EEEE or RRRR for bispecific exchange as describe in Example 8.
  • Total bone marrow mononuclear cells from myeloma patients were seeded in clear U-bottom plates at total bone marrow mononuclear cell numbers that resulted in 3000-5000 myeloma cells/well. Cells were treated with 10-fold serially diluted bispecific antibody. Five days after treatment, total viable cells were determined by flow cytometry using antibodies to CD138 and CD38 (Biolegend, CA). Cells were incubated with antibodies at 4° in PBS+0.5% FIBS for 30 minutes. Cells were washed and Fixable Viability Dye eFluor 780 (eBioscience, Inc., CA) in PBS was added to the cells for 30 minutes at 4°. Prior to cell acquisition on a BD flow cytometer, cells were washed and CountBright Absolute Counting Beads (Molecular Probes, OR) were added. Percent live cells were determined as live cell count in treated vs untreated wells using counting beads. EC50 was calculated by Prism software.
  • Table 13A shows that all human anti-BCMA_h2B4 bispecific antibodies have cell killing activity on myeloma patient samples and patient T cells are functional effector cells. Table 13B shows killing of one anti-BCMA bispecific on multiple myeloma patient samples with different effector to target (E:T) ratio.
  • TABLE 13A
    Anti-BCMA bispecific
    (Patient MM00146) EC50 (nM)
    P6E01 L3PY/H3TAQ 0.015
    P5A2 0.946
    A02_Rd4_0.6 nM_C01 0.064
    A02_Rd4_6 nM_C16 0.029
    NNC_2b41d hIgG2dA 3.302
  • TABLE 13B
    Combo_Rd4_0.6 nM_C29
    Patient EC50 (nM) E:T
    MM00146 0.035 1:1
    MM00147 0.031 2:1
    MM00151 0.02 3:1
    MM00152 0.289 1:2
  • Example 17: ELISPOT of Antibody Secreting Cells from Cynomolgus Monkeys Administered with Anti-BCMA/CD3 Bispecific Antibodies
  • This example illustrates the depletion of IgG secreting cells in cynomolgus monkey with anti-BCMA/CD3 bispecific antibodies.
  • Cynomolgus monkeys (n=2) were dosed via intravenous bolus injection with bispecific anti-BCMA_CD3 antibodies (h2B4-VH-hnps VL_TK), A02_Rd4_0.6 nM_C01/H2B4 and Combo_Rd4_0.6 nM_C29/H2B at two doses, day 1 and day 8, of 100 ug/kg and 300 ug/kg. Animals were observed twice daily and at each blood collection time point. Peripheral blood mononuclear cells (PBMC) were sampled on days −6, 4 and 10. Bone marrow samples were taken on day 10 when animals were necropsied.
  • Blood was collected into Becton Dickinson® CPT™ Cell Preparation Tubes containing sodium heparin and a density gradient, and then PBMCs were collected at the gradient interface following centrifugation.
  • A sample of bone marrow (from femur) was collected by flushing with approximately 5 mL of 100% fetal bovine serum (FBS) and then single cell suspensions were prepared by suspending flushed marrow in 50 ml of buffer.
  • Cells were counted using the Cellometer Vision and adjusted with complete RPMI 1640 culture medium to a concentration of 5×106 cells per mL for PBMC and 2×106 cells per mL for bone marrow cells. Total IgG-secreting cells were enumerated using the ELISpotBASIC kit from Mabtech (#3850-2HW-Plus). Briefly, PBMC or bone marrow cells were added to triplicate wells at specified concentrations (PBMCs at 5×105/well, and bone marrow cells at 2×105/well), and then cells were serially diluted in the plate. After an overnight incubation, plates were washed and a biotinylated detection antibody was added. Plates were incubated for 2 hours and streptavidin-HRP was added for 1 hour. IgG spots were visualized using TMB substrate solution and counted using the ImmunoSpot Imaging Analyzer system (CTL) and ImmunoSpot 5.1 software. Data were expressed as the mean (+/−SD) number of IgG-secreting cells from triplicate samples.
  • The resulting IgG-secreting cell count in PBMC and in bone marrow are listed in Tables 14A and 14B, respectively. Depletion of IgG-secreting cells was seen for both anti-BCMA/CD3 bispecific antibodies in PBMC as compared to pre-dose and bone marrow as compared to vehicle and negative control. A dose dependent effect was seen in the bone marrow.
  • TABLE 14A
    Day
    PBMC −6 4 10
    Vehicle 204, 320 323, 538 127, 137
    0.1 mg/kg Combo_Rd4_0.6 107, 128 5, 0 0, 0
    nM_C29
    0.3 mg/kg Combo_Rd4_0.6 304, 362 0, 5 0, 0
    nM_C29
    0.1 mg/kg A02_Rd4_0.6 447, 672 16, 27 21, 10
    nM_C01
    0.3 mg/kg A02_Rd4_0.6 512, 224 25, 8 0, 0
    nM_C01
    0.3 mg/kg NNC_2b41d 139, 361 1345, 1154 992, 928
  • Example 18: Anti-BCMA/CD3 Bispecific Induce Tumor Regression and Inhibition in MM1.S Tumor Model
  • TABLE 14B
    Bone Marrow Day 10
    Vehicle 2614, 8093
    0.1 mg/kg Combo_Rd4_0.6 nM_C29 35, 18
    0.3 mg/kg Combo_Rd4_0.6 nM_C29 35, 22
    0.1 mg/kg A02_Rd4_0.6 nM_C01 996, 960
    0.3 mg/kg A02_Rd4_0.6 nM_C01 2170, 93 
    0.3 mg/kg NNC_2b41d 5980, 2893
  • This example illustrates tumor regression and inhibition in an orthotopic MM1.S myeloma model.
  • In vivo efficacy study of BCMA bispecifics was performed with MM1.S, expressing luciferase and GFP, orthotopic model. One day prior to tumor cell injection, mice were irradiated with 100cGy using RS 2000 Biological Research Irradiator (RAD Source Technolgies, GA). Five million MM1.S LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female Nod/Scid/IL2Rg−/− (NSG) animals. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enabled monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 15E6 for all animals, the animals were injected through bolus tail vein with 20 million expanded T cells from PBMC. Briefly, pan-T cells purchased from AllCells (Alameda, CA) were activated with human T Cell Activation/Expansion Kit (Miltenyi, San Diego, CA). After three days, 15U/mL of IL2 (ebioscience, San Diego, CA) was added every two days until day 11. Cells were harvested, activation/expansion beads were magnetically removed, and cells were washed and resuspended in PBS. One day post T cell injection, mice were imaged as described above and animals were randomized into groups of seven mice; A02_Rd4_0.6 nM_C01 at 0.03 mg/kg and 0.3 mg/kg and Combo_Rd4_0.6 nM_C29 at 0.03 mg/kg, 0.1 mg/kg and 0.3 mg/kg. A single dose of human anti-BCMA/CD3 (h2B4-VH-wt VL_TK) bispecific and negative (NNC) control bispecific antibody was administered through bolus tail vein injection. Animals were sacrificed when they exhibited hindlimb paralysis, an endpoint for MM1.S orthotopic model. FIG. 12 shows that a single dose of human anti-BCMA/CD3 bispecific antibody resulted in tumor regression in a dose-dependent manner.
  • Example 19: Two Doses of Anti-BCMA/CD3 Bispecific Induce Tumor Regression in Aggressive Molp8 Tumor Model
  • This example illustrates tumor regression with two doses of anti-BCMA/CD3 bispecific antibodies in an orthotopic Molp8 myeloma model.
  • In vivo efficacy study of BCMA bispecifics was performed with Molp8, expressing luciferase and GFP, orthotopic model. Two million Molp8 LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female NSG animals. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enabled monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). When the total flux reached an average of 25E6 for all animals were randomized into three groups of seven mice; 1) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg, 2) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg, two doses and 3) NNC_2B4, 0.3 mg/kg, two doses. The animals were injected through bolus tail vein with 20 million expanded T cells as described in Example 18. Two days post T cell injection, mice were dosed with bispecific antibodies. Animals were sacrificed when they exhibited weight loss of more than 15%, an endpoint for Molp8 orthotopic model. FIG. 13 shows that two doses of human anti-BCMA/CD3 (h2B4-VH-wt VL_TK) bispecific antibody resulted in increased tumor regression.
  • Example 20: Anti-BCMA/CD3 Bispecific in Combination with Standard of Care for Multiple Myeloma in Orthotopic Molp8 Tumor Model
  • This example demonstrates no opposing effects on anti-BCMA/CD3 bispecific antibodies when combined with bortezomib or lenalidomide and better potency with anti-BCMA/CD3 bispecific antibodies as compared to bortezomib and lenalidomie combined.
  • In vivo efficacy study of BCMA bispecifics was performed with Molp8, expressing luciferase and GFP, orthotopic model. Two million Molp8 LucGFP cells were injected intravenously through the tail vein into 6-8 weeks old female NSG animals. Intraperitoneal injection of D-luciferin (Regis Technologies, Morton Grove, IL) (200 uL per animal at 15 mg/mL), followed by anesthesia with isofluorane and subsequent whole body bioluminescence imaging (BLI) enabled monitoring of tumor burden. Bioluminescent signals emitted by the interaction between luciferase expressed by the tumor cells and luciferin were captured by imaging using an IVIS Spectrum CT (Perkin Elmer, MA) and quantified as total flux (photons/sec) using Living Image 4.4 (Caliper Life Sciences, Alameda, CA). The animals were injected through bolus tail vein on day 7 with 20 million expanded T cells as described in Example 18. Two days post T cell injection, mice were imaged and randomized into five groups of seven mice with an average of 17E6 total flux/group: 1) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg, 2) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg and 1 mg/kg bortezomib, 3) Combo_Rd4_0.6 nM_C29, 0.3 mg/kg and 50 mg/kg lenalidomide, 4) 1 mg/kg bortezomib and 50 mg/kg lenalidomide and 5) vehicle. Anti-BCMA/CD3 (in PBS) was injected through bolus tail vein injection, bortezomib (in PBS) was administrated via intraperitoneal injection and lenalidomide (30% PEG400/5% propylene glycol/0.5% Tween80) via oral gavage. Vehicle consisted of 30% PEG400/5% propylene glycol/0.5% Tween80, which was administrated via oral gavage. Animals were sacrificed when they exhibit weight loss of more than 15%, an endpoint for Molp8 orthotopic model. FIG. 14 shows that combining an anti-BCMA/CD3 (h2B4-VH-hnps VL-TK) bispecific antibody with bortezomib or lenalidomide did not have a negative effect on the efficacy of the anti-BCMA/CD3 bispecific antibody. In this model, anti-BCMA/CD3 bispecific antibody alone or in combination with lenalidomide or bortezomib is more efficacious than lenalidomide and bortezomib combined.
  • Example 21: In Vitro Study of Anti-BCMA/CD3 Bispecific in Combination with Lenalidomide, Carfilzomib or Doxorubicin on OPM2 Cell Line
  • This example illustrates no adverse effects on T cell function when combined with carfilzomib, doxorubicin, and lenalidomide for anti-BCMA/CD3 bispecific antibody activity as compared to the bispecific antibody alone.
  • CD3+ T cells from PBMC were negatively selected using Pan T Cell Isolation kit, human (Miltenyi, San Diego CA). OPM2 cells and CD3+ T-cells were seeded in clear U-bottom plates 20000 and 100000 cells/well, respectively. OPM2 and CD3+ T cells were first incubated with the standard of care for two hours at 37°. 1.56 nM carfilzomib and 6.25 nM doxorubicin were diluted in PBS containing 0.02% DMSO. Lenalidomide was diluted in PBS containing 0.1% DMSO at 195 nM. Cells were treated with 10-fold serially diluted bispecific antibody. Three days after treatment, total viable cells were determined by flow cytometry using antibodies to CD138, CD4, and CD8 (Biolegend). Cells were incubated with antibodies at 4° in PBS+0.5% FBS for 30 minutes. Cells were washed and Fixable Viability Dye eFluor 780 (eBioscience, Inc., CA) in PBS was added to the cells for 30 minutes at 4°. Prior to cell acquisition on a BD flow cytometer, cells were washed and CountBright Absolute Counting Beads (Molecular Probes, OR) were added. Percent live cells were determined as live cell count in treated vs untreated wells using counting beads. FIGS. 15A, 15B, and 15C, respectively, show that carfilzomib, lenalidomide, and doxorubicin do not have a negative effect on the function of Combo_Rd4_0.6 nM_C29-CD3 (h2B4-VH-hnps VL-TK) bispecific antibody on OPM2 cells.
  • Example 22: In Vitro Study of Anti-BCMA/CD3 Bispecific in Combination with Lenalidomide and Carfilzomib on KMS12BM Cell Line
  • This example illustrates synergistic effects on anti-BCMA/CD3 bispecific function when combined with carfilzomib and lenalidomide as compared to each molecule alone.
  • CD3+ T cells from PBMC were negatively selected using Pan T Cell Isolation kit, human (Miltenyi, San Diego CA). KMS12BM cells and CD3+ T-cells were seeded in clear U-bottom plates 20000 and 100000 cells/well, respectively. Cells were treated with 0.017 nM anti-BCMA/CD3 bispecific in combination with carfilzomib and a range of concentration for lenalidomide. 1.25 nM carfilzomib was diluted in PBS containing 0.02% DMSO. Lenalidomide was diluted in PBS containing 0.1% DMSO starting at 4 uM diluted 4-fold. Three days after treatment, total viable cells were determined by flow cytometry using antibodies to CD138, CD4, and CD8 (Biolegend, CA). Cells were incubated with antibodies at 4° in PBS+0.5% FBS for 30 minutes. Cells were washed and Fixable Viability Dye eFluor 780 (eBioscience, Inc., CA) in PBS was added to the cells for 30 minutes at 4°. Prior to cell acquisition on a BD flow cytometer, cells were washed and CountBright Absolute Counting Beads (Molecular Probes, OR) were added. Percent live cells were determined as live cell count in treated versus untreated wells using counting beads. FIG. 16 shows that at the concentrations tested, carfilzomib, lenalidomide, and anti-BCMA/CD3 (h2B4-VH-hnps VL-TK) bispecific antibody had very little single agent cytotoxic function. When all three agents were combined, a synergistic effect was observed at a dose-dependent lenalidomide concentration in KMS12BM cells.
  • Although the disclosed teachings have been described with reference to various applications, methods, kits, and compositions, it will be appreciated that various changes and modifications can be made without departing from the teachings herein and the claimed invention below. The foregoing examples are provided to better illustrate the disclosed teachings and are not intended to limit the scope of the teachings presented herein. While the present teachings have been described in terms of these exemplary embodiments, the skilled artisan will readily understand that numerous variations and modifications of these exemplary embodiments are possible without undue experimentation. All such variations and modifications are within the scope of the current teachings.
  • All references cited herein, including patents, patent applications, papers, text books, and the like, and the references cited therein, to the extent that they are not already, are hereby incorporated by reference in their entirety. In the event that one or more of the incorporated literature and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls.
  • The foregoing description and Examples detail certain specific embodiments of the invention and describes the best mode contemplated by the inventors. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the invention may be practiced in many ways and the invention should be construed in accordance with the appended claims and any equivalents thereof.

Claims (45)

It is claimed:
1. An isolated antibody, or an antigen binding fragment thereof, which specifically binds to B-Cell Maturation Antigen (BCMA), wherein the antibody comprises
(a) a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence SYX1MX2, wherein X1 is A or P; and X2 is T, N, or S (SEQ ID NO: 301), GFTFX1SY, wherein X1 is G or S (SEQ ID NO: 302), or GFTFX1SYX2MX3, wherein X1 is G or S, X2 is A or P; and X3 is T, N, or S (SEQ ID NO: 303); (ii) a VH CDR2 comprising the sequence AX1X2X3X4GX5X6X7X8YADX9X10KG, wherein X1 is I, V, T, H, L, A, or C; X2 is S, D, G, T, I, L, F, M, or V; X3 is G, Y, L, H, D, A, S, or M; X4 is S, Q, T, A, F, or W; X5 is G or T; X6 is N, S, P, Y, W, or F; X7 is S, T, I, L, T, A, R, V, K, G, or C; X8 is F, Y, P, W, H, or G; X9 is V, R, or L; and X10 is G or T (SEQ ID NO: 305), or X1X2X3X4X5X6, wherein X1 is S, V, I, D, G, T, L, F, or M; X2 is G, Y, L, H, D, A, S, or M; X3 is S, G, F, or W; X4 is G or S; X5 is G or T; and X6 is N, S, P, Y, or W (SEQ ID NO: 306); and iii) a VH CDR3 comprising the sequence VSPIX1X2X3X4, wherein X1 is A or Y; X2 is A or S; and X3 is G, Q, L, P, or E (SEQ ID NO: 307), or YWPMX1X2, wherein X1 is D, S, T, or A; and X2 is I, S, L, P, or D (SEQ ID NO: 308); and/or
(b) a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence X1X2X3X4X5X6X7X8X9X10X11X12, wherein X1 is R, G, W, A, or C; X2 is A, P, G, L, C, or S; X3 is S, G, or R; X4 is Q, C, E, V, or I; X5 is S, P, G, A, R, or D; X6 is V, G, I, or L; X7 is S, E, D, P, or G; X8 is S, P, F, A, M, E, V, N, D, or Y; X9 is I, T, V, E, S, A, M, Q, Y, H, R, or F; X10 is Y or F; X11 is L, W, or P; and X12 is A, S, or G (SEQ ID NO: 309); (ii) a VL CDR2 comprising the sequence X1ASX2RAX3, wherein X1 is G or D; X2 is S or I; and X3 is T or P (SEQ ID NO: 310); and (iii) a VL CDR3 comprising the sequence QQYX1X2X3PX4T, wherein X1 is G, Q, E, L, F, A, S, M, K, R, or Y; X2 is S, R, T, G, V, F, Y, D, A, H, V, E, K, or C; X3 is W, F, or S; and X4 is L or I (SEQ ID NO: 311), or QQYX1X2X3PX4, wherein X1 is G, Q, E, L, F, A, S, M, R, K, or Y; X2 is S, R, T, G, R, V, D, A, H, E, K, C, F, or Y; X3 is W, S, or F; and X4 is L or I (SEQ ID NO: 312).
2. An isolated antibody, or an antigen binding fragment thereof, which specifically binds to BCMA, wherein the antibody comprises:
a VH region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 2, 3, 7, 8, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35, 37, 39, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 83, 87, 92, 95, 97, 99, 101, 104, 106, 110, 112, 114, 118, 120, 122, 125, 127, 313, 314, 363, or 365; and/or
a VL region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 1, 4, 5, 6, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 34, 36, 38, 40, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 317, 80, 81, 82, 84, 85, 86, 88, 89, 90, 91, 93, 94, 96, 98,100, 102, 103, 105, 107, 108, 109, 111, 113, 115, 116, 117, 119, 121, 123, 124, 126, 128, 315, 316, or 364.
3. The antibody or the antigen binding fragment of claim 2, wherein the VH region comprises (i) a VH CDR1 comprising SEQ ID NO:150, 151, 152, 156, or 157; (ii) a VH CDR2 comprising SEQ ID NO: 169, 154, 194, 159, 195, 196, 162, 158, 198, 177, 178, 199, 200, 201, 202, 203, 204, 206, 207, 208, 172, 203, or 204; and (iii) a VH CDR3 comprising SEQ ID NO: 155, 161, 197, 205, or 164; and/or wherein the VL region comprises (i) a VL CDR1 comprising SEQ ID NO: 209, 271, 273, 275, 251, 277, 260, 279, 245, 283, 285, 287, 290, 292, 235, 297, or 299; (ii) a VL CDR2 comprising SEQ ID NO: 221; and (iii) a VL CDR3 comprising SEQ ID NO: 225, 272, 274, 276, 278, 280, 281, 282, 284, 286, 288, 289, 291, 293, 294, 229, 296, 298, or 300.
4. The antibody or the antigen binding fragment of claim 3, wherein the VH region comprises (i) a VH CDR1 comprising SEQ ID NO: 151, 156, or 157; (ii) a VH CDR2 comprising SEQ ID NO: 158 or 159; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and/or wherein the VL region comprises (i) a VL CDR1 comprising SEQ ID NO: 209; (ii) a VL CDR2 comprising SEQ ID NO: 221; and (iii) a VL CDR3 comprising SEQ ID NO: 225.
5. The antibody or the antigen binding fragment of claim 4, wherein the VH region comprises the sequence shown in SEQ ID NO: 112 or a variant with one or several conservative amino acid substitutions in residues that are not within a CDR and/or the VL region comprises the amino acid sequence shown in SEQ ID NO: 38 or a variant thereof with one or several amino acid substitutions in amino acids that are not within a CDR.
6. The antibody or the antigen binding fragment of claim 5, wherein the antibody comprises a light chain comprising the sequence shown in SEQ ID NO: 357 and a heavy chain comprising the sequence shown in SEQ ID NO: 358.
7. The antibody or the antigen binding fragment of claim 3, wherein the antibody comprises a VH region produced by the expression vector with ATCC Accession No. PTA-122094.
8. The antibody or the antigen binding fragment of claim 4, wherein the antibody comprises a VL region produced by the expression vector with ATCC Accession No. PTA-122093.
9. An isolated antibody which specifically binds to BCMA and competes with the antibody of claim 4.
10. A pharmaceutical composition comprising a therapeutically effective amount of the antibody or antigen binding fragment of claim 1 and a pharmaceutically acceptable carrier.
11. An isolated polynucleotide comprising a nucleotide sequence encoding the antibody of claim 1.
12. A vector comprising the polynucleotide of claim 11.
13. An isolated host cell that recombinantly produces the antibody or the antigen binding fragment of claim 1.
14. A method of producing an antibody, comprising culturing the host cell of claim 13 under conditions that result in production of the antibody, and isolating the antibody from the host cell or culture.
15. A method of treating a condition associated with cells expressing BCMA in a subject comprising administering to a subject in need thereof an effective amount of the pharmaceutical composition of claim 10.
16. The method of claim 15, wherein the condition is a cancer.
17. The method of claim 16, wherein the cancer is a B-cell related cancer selecting from the group consisting of multiple myeloma, malignant plasma cell neoplasm, Hodgkin's lymphoma, nodular lymphocyte predominant Hodgkin's lymphoma, Kahler's disease and Myelomatosis, plasma cell leukemia, plasmacytoma, B-cell prolymphocytic leukemia, hairy cell leukemia, B-cell non-Hodgkin's lymphoma (NHL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), follicular lymphoma, Burkitt's lymphoma, marginal zone lymphoma, mantle cell lymphoma, large cell lymphoma, precursor B-lymphoblastic lymphoma, myeloid leukemia, Waldenstrom's macroglobulienemia, diffuse large B cell lymphoma, follicular lymphoma, marginal zone lymphoma, mucosa-associated lymphatic tissue lymphoma, small cell lymphocytic lymphoma, mantle cell lymphoma, Burkitt lymphoma, primary mediastinal (thymic) large B-cell lymphoma, lymphoplasmactyic lymphoma, Waldenström macroglobulinemia, nodal marginal zone B cell lymphoma, splenic marginal zone lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, lymphomatoid granulomatosis, T cell/histiocyte-rich large B-cell lymphoma, primary central nervous system lymphoma, primary cutaneous diffuse large B-cell lymphoma (leg type), EBV positive diffuse large B-cell lymphoma of the elderly, diffuse large B-cell lymphoma associated with inflammation, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, large B-cell lymphoma arising in HHV8-associated multicentric Castleman disease, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma, and other B-cell related lymphoma.
18. An isolated antibody, or an antigen binding fragment thereof, which specifically binds to CD3, wherein the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443, 445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399.
19. The antibody or the antigen binding fragment of claim 18, wherein the antibody comprises a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387.
20. The antibody or the antigen binding fragment of claim 18, wherein the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 340, 343, 430, 431, 435, or 440, 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341, 433, 452, or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342, 432, 434, 437, 438, 439, 446, or 453.
21. The antibody or the antigen binding fragment of claim 20, wherein the VH region comprises (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 407, or 408 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336, 404, 405, or 417; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335 or 406; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343 or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341 or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342 or 439.
22. An isolated antibody which specifically binds to CD3 and competes with the antibody of claim 21.
23. An isolated polynucleotide comprising a nucleotide sequence encoding the antibody of claim 18.
24. A vector comprising the polynucleotide of claim 23.
25. An isolated host cell that recombinantly produces the antibody or the antigen binding fragment of claim 18.
26. A method of producing an antibody, comprising culturing the host cell of claim 25 under conditions that result in production of the antibody, and isolating the antibody from the host cell or culture.
27. A bispecific antibody wherein the bispecific antibody is a full-length human antibody, comprising a first antibody variable domain of the bispecific antibody capable of recruiting the activity of a human immune effector cell by specifically binding to an effector antigen located on the human immune effector cell, and comprising a second antibody variable domain of the bispecific antibody capable of specifically binding to a target antigen, wherein the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 320, 322, 324, 326, 328, 330, 345, 347, 349, 351, 444, 354, 356, 378, 442, 380, 382, 384 386, 388, 390, 392, 394, 396, 398, or 400; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 319, 321, 323, 325, 327, 329, 344, 346, 348, 350, 352, 355, 377, 443, 445, 379, 381, 383, 385, 387, 389, 391, 393, 395, 397, or 399.
28. The bispecific antibody of claim 27, wherein the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: SEQ ID NO: 331, 332, 333, 401, 402, 403, 407, 408, 415, 416, 418, 419, 420, 424, 425, 426, 446, 447, or 448 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 334, 336, 337, 338, 339, 404, 405, 409, 410, 411, 412, 413, 414, 417, 418, 421, 422, 427, 428, 449, or 450; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335, 406, 423, 429, or 451; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 340, 343, 430, 431, 435, 440, or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341, 433, 452, or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342, 432, 434, 437, 438, 439, 446, or 453.
29. The bispecific antibody of claim 28, wherein the first antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 324 or 388; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 323 or 387; and the second antibody variable domain comprises a heavy chain variable (VH) region comprising a VH CDR1, VH CDR2, and VH CDR3 of the VH sequence shown in SEQ ID NO: 112; and/or a light chain variable (VL) region comprising VL CDR1, VL CDR2, and VL CDR3 of the VL sequence shown in SEQ ID NO: 38.
30. The bispecific antibody of claim 27, wherein the second antibody variable domain comprises
a. a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence SYX1MX2, wherein X1 is A or P; and X2 is T, N, or S (SEQ ID NO: 301), GFTFX1SY, wherein X1 is G or S (SEQ ID NO: 302), or GFTFX1SYX2MX3, wherein X1 is G or S, X2 is A or P; and X3 is T, N, or S (SEQ ID NO: 303); (ii) a VH CDR2 comprising the sequence AX1X2X3X4GX5X6X7X8YADX9X10KG, wherein X1 is I, V, T, H, L, A, or C; X2 is S, D, G, T, I, L, F, M, or V; X3 is G, Y, L, H, D, A, S, or M; X4 is S, Q, T, A, F, or W; X5 is G or T; X6 is N, S, P, Y, W, or F; X7 is S, T, I, L, T, A, R, V, K, G, or C; X8 is F, Y, P, W, H, or G; X9 is V, R, or L; and X10 is G or T (SEQ ID NO: 305), or X1X2X3X4X5X6, wherein X1 is S, V, I, D, G, T, L, F, or M; X2 is G, Y, L, H, D, A, S, or M; X3 is S, G, F, or W; X4 is G or S; X5 is G or T; and X6 is N, S, P, Y, or W (SEQ ID NO: 306); and iii) a VH CDR3 comprising the sequence VSPIX1X2X3X4 wherein X1 is A or Y; X2 is A or S; and X3 is G, Q, L, P, or E (SEQ ID NO: 307), or YWPMX1X2, wherein X1 is D, S, T, or A; and X2 is I, S, L, P, or D (SEQ ID NO: 308); and/or
b. a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence X1X2X3X4X5X6X7X8X9X10X11X12, wherein X1 is R, G, W, A, or C; X2 is A, P, G, L, C, or S; X3 is S, G, or R; X4 is Q, C, E, V, or I; X5 is S, L, P, G, A, R, or D; X6 is V, G, or I; X7 is S, E, D, or P; X8 is S, P, F, A, M, E, V, N, D, or Y; X9 is I, T, V, E, S, A, M, Q, Y, H, or R; X10 is Y or F; X11 is L, W, or P; and X12 is A, S, or G (SEQ ID NO: 309); (ii) a VL CDR2 comprising the sequence X1ASX2RAX3, wherein X1 is G or D; X2 is S or I; and X3 is T or P (SEQ ID NO: 310); and (iii) a VL CDR3 comprising the sequence QQYX1X2X3PX4T, wherein X1 is G, Q, E, L, F, A, S, M, K, R, or Y; X2 is S, R, T, G, V, F, Y, D, A, H, V, E, K, or C; X3 is W, F, or S; and X4 is L or I (SEQ ID NO: 311), or QQYX1X2X3PX4, wherein X1 is G, Q, E, L, F, A, S, M, R, K, or Y; X2 is S, R, T, G, R, V, D, A, H, E, K, C, F, or Y; X3 is W, S, or F; and X4 is L or I (SEQ ID NO: 312).
31. The bispecific antibody of claim 27, wherein the second antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO:150, 151, 152, 156, 157, 348, 349, 353, 354, or 355; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 169, 154, 194, 159, 195, 196, 162, 158, 198, 177, 178, 199, 200, 201, 202, 203, 204, 206, 207, 208, 172, 203, 204, 350, 351, 356 or 357; and (iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 155, 161, 197, 205,164, or 352, or 358; and/or wherein the light chain variable (VL) region comprises (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209, 271, 273, 275, 251, 277, 260, 279, 245, 283, 285, 287, 290, 292, 235, 297, 299, or 361; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221, 359 or 362; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 211, 225, 272, 274, 276, 278, 280, 281, 282, 284, 286, 288, 289, 291, 293, 294, 229, 296, 298, 300 or 360.
32. The bispecific antibody of claim 31, wherein the
a. first antibody variable domain comprises a heavy chain variable (VH) region comprising (i) a VH complementarity determining region one (CDR1) comprising the sequence shown in SEQ ID NO: 331, 332, 333, 401, 407, or 408 (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 336, 417, 404, or 405; and iii) a VH CDR3 comprising the sequence shown in SEQ ID NO: 335 or 406; and/or a light chain variable (VL) region comprising (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 343 or 441; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 341 or 436; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 342 or 439; and the
b. the second antibody variable domain comprises a heavy chain VH region comprising a heavy chain variable (VH) region comprising (i) a VH CDR1 comprising the sequence shown in SEQ ID NO: 151, 156, or 157; (ii) a VH CDR2 comprising the sequence shown in SEQ ID NO: 158 OR 159; and (iii) a VH CDR3 comprising SEQ ID NO: 155; and/or wherein the light chain variable (VL) region comprises (i) a VL CDR1 comprising the sequence shown in SEQ ID NO: 209; (ii) a VL CDR2 comprising the sequence shown in SEQ ID NO: 221; and (iii) a VL CDR3 comprising the sequence shown in SEQ ID NO: 225.
33. The bispecific antibody of claim 27, wherein both the first and the second antibody variable domains of the heterodimeric protein comprise amino acid modifications at positions 223, 225, and 228 in the hinge region and at position 409 or 368 (EU numbering scheme) in the CH3 region of a human IgG2 (SEQ ID NO: 493).
34. The bispecific antibody of claim 33, further comprising an amino acid modification at position 265 of the human IgG2.
35. A nucleic acid encoding the antibody of claim 27.
36. A vector comprising the nucleic acid of claim 35.
37. A host cell comprising the nucleic acid of claim 35 or the vector of claim 36.
38. A pharmaceutical composition comprising the bispecific antibody of claim 27.
39. A method of treating a condition associated with malignant cells expressing a tumor antigen in a subject comprising administering to the subject in need thereof an effective amount of the pharmaceutical composition of claim 38.
40. The method of claim 39, wherein the condition is a cancer.
41. The method of claim 40, wherein the cancer is a B-cell related cancer selecting from the group consisting of multiple myeloma, malignant plasma cell neoplasm, Hodgkin's lymphoma, nodular lymphocyte predominant Hodgkin's lymphoma, Kahler's disease and Myelomatosis, plasma cell leukemia, plasmacytoma, B-cell prolymphocytic leukemia, hairy cell leukemia, B-cell non-Hodgkin's lymphoma (NHL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), follicular lymphoma, Burkitt's lymphoma, marginal zone lymphoma, mantle cell lymphoma, large cell lymphoma, precursor B-lymphoblastic lymphoma, myeloid leukemia, Waldenstrom's macroglobulienemia, diffuse large B cell lymphoma, follicular lymphoma, marginal zone lymphoma, mucosa-associated lymphatic tissue lymphoma, small cell lymphocytic lymphoma, mantle cell lymphoma, Burkitt lymphoma, primary mediastinal (thymic) large B-cell lymphoma, lymphoplasmactyic lymphoma, Waldenström macroglobulinemia, nodal marginal zone B cell lymphoma, splenic marginal zone lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, lymphomatoid granulomatosis, T cell/histiocyte-rich large B-cell lymphoma, primary central nervous system lymphoma, primary cutaneous diffuse large B-cell lymphoma (leg type), EBV positive diffuse large B-cell lymphoma of the elderly, diffuse large B-cell lymphoma associated with inflammation, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, large B-cell lymphoma arising in HHV8-associated multicentric Castleman disease, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma, B-cell lymphoma unclassified with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma, and other B-cell related lymphoma.
42. A method of inhibiting tumor growth or progression in a subject who has malignant cells expressing BCMA, comprising administering to the subject in need thereof an effective amount of the pharmaceutical composition comprising the bispecific antibody of claim 30 to the subject.
43. A method of inhibiting metastasis of malignant cells expressing BCMA in a subject, comprising administering to the subject in need thereof an effective amount of the pharmaceutical composition comprising the bispecific antibody of claim 30 to the subject.
44. A method of inducing tumor regression in a subject who has malignant cells expressing BCMA, comprising administering to the subject in need thereof an effective amount of the pharmaceutical composition comprising the bispecific antibody of claim 30 to the subject.
45. A method of treating multiple myeloma in a subject in need thereof, comprising administering to the subject an effective amount of the pharmaceutical composition comprising the bispecific antibody of claim 30 to the subject and one or more other therapeutic agent selecting from the group consisting of bortezomib, lenalidomide, carfilzomib, and doxorubicin.
US18/456,399 2015-04-13 2023-08-25 Therapeutic antibodies and their uses Pending US20240018252A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/456,399 US20240018252A1 (en) 2015-04-13 2023-08-25 Therapeutic antibodies and their uses

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201562146843P 2015-04-13 2015-04-13
US201562146504P 2015-04-13 2015-04-13
US201662301582P 2016-02-29 2016-02-29
US15/085,644 US9969809B2 (en) 2015-04-13 2016-03-30 Therapeutic antibodies and their uses
US15/878,344 US10040860B2 (en) 2015-04-13 2018-01-23 Therapeutic antibodies and their uses
US16/017,974 US10793635B2 (en) 2015-04-13 2018-06-25 Therapeutic antibodies and their uses
US17/005,073 US11814435B2 (en) 2015-04-13 2020-08-27 Therapeutic antibodies and their uses
US18/456,399 US20240018252A1 (en) 2015-04-13 2023-08-25 Therapeutic antibodies and their uses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US17/005,073 Continuation US11814435B2 (en) 2015-04-13 2020-08-27 Therapeutic antibodies and their uses

Publications (1)

Publication Number Publication Date
US20240018252A1 true US20240018252A1 (en) 2024-01-18

Family

ID=55661497

Family Applications (6)

Application Number Title Priority Date Filing Date
US15/085,644 Active 2036-07-08 US9969809B2 (en) 2015-04-13 2016-03-30 Therapeutic antibodies and their uses
US15/878,344 Active US10040860B2 (en) 2015-04-13 2018-01-23 Therapeutic antibodies and their uses
US16/017,974 Active US10793635B2 (en) 2015-04-13 2018-06-25 Therapeutic antibodies and their uses
US17/005,073 Active 2037-01-15 US11814435B2 (en) 2015-04-13 2020-08-27 Therapeutic antibodies and their uses
US17/129,833 Active US11155630B2 (en) 2015-04-13 2020-12-21 Therapeutic antibodies and their uses
US18/456,399 Pending US20240018252A1 (en) 2015-04-13 2023-08-25 Therapeutic antibodies and their uses

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US15/085,644 Active 2036-07-08 US9969809B2 (en) 2015-04-13 2016-03-30 Therapeutic antibodies and their uses
US15/878,344 Active US10040860B2 (en) 2015-04-13 2018-01-23 Therapeutic antibodies and their uses
US16/017,974 Active US10793635B2 (en) 2015-04-13 2018-06-25 Therapeutic antibodies and their uses
US17/005,073 Active 2037-01-15 US11814435B2 (en) 2015-04-13 2020-08-27 Therapeutic antibodies and their uses
US17/129,833 Active US11155630B2 (en) 2015-04-13 2020-12-21 Therapeutic antibodies and their uses

Country Status (28)

Country Link
US (6) US9969809B2 (en)
EP (3) EP4234581A3 (en)
JP (3) JP6913025B2 (en)
KR (3) KR102110557B1 (en)
CN (3) CN114773476A (en)
AU (3) AU2016248946B2 (en)
BR (1) BR112017019785B1 (en)
CA (4) CA3219684A1 (en)
CO (1) CO2017010495A2 (en)
DK (1) DK3283106T3 (en)
ES (1) ES2905315T3 (en)
FI (1) FIC20240014I1 (en)
HK (1) HK1255385A1 (en)
HU (1) HUE057285T2 (en)
IL (4) IL310723A (en)
MX (3) MX2017013297A (en)
MY (1) MY196625A (en)
PE (3) PE20180046A1 (en)
PH (1) PH12017501732A1 (en)
PL (1) PL3283106T3 (en)
PT (1) PT3283106T (en)
RU (1) RU2722381C2 (en)
SA (1) SA517390140B1 (en)
SG (3) SG10201912823PA (en)
SI (1) SI3283106T1 (en)
TW (3) TWI703159B (en)
WO (1) WO2016166629A1 (en)
ZA (1) ZA201807153B (en)

Families Citing this family (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112017019914A2 (en) 2015-04-13 2018-06-19 Pfizer b-maturation antigen-directed chimeric antigen receptors
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
US10221242B2 (en) 2016-01-21 2019-03-05 Pfizer Inc. Antibodies specific for epidermal growth factor receptor variant III and their uses
SG11201806261XA (en) 2016-02-17 2018-09-27 Seattle Genetics Inc Bcma antibodies and use of same to treat cancer and immunological disorders
CN109328074A (en) 2016-04-01 2019-02-12 凯德药业股份有限公司 Chimeric antigen and T cell receptor and the method used
CR20210084A (en) 2016-04-01 2021-03-26 Amgen Inc Chimeric receptors and methods of use thereof
CN117903307A (en) 2016-04-01 2024-04-19 凯德药业股份有限公司 BCMA binding molecules and methods of use thereof
WO2017189959A1 (en) 2016-04-29 2017-11-02 Voyager Therapeutics, Inc. Compositions for the treatment of disease
EP3448987A4 (en) 2016-04-29 2020-05-27 Voyager Therapeutics, Inc. Compositions for the treatment of disease
WO2018035710A1 (en) * 2016-08-23 2018-03-01 Akeso Biopharma, Inc. Anti-ctla4 antibodies
JP2020506700A (en) 2017-01-31 2020-03-05 ノバルティス アーゲー Cancer treatment using multispecific chimeric T cell receptor protein
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
JOP20190247A1 (en) * 2017-04-20 2019-10-20 Lilly Co Eli ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
US20200055948A1 (en) 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
EP3615068A1 (en) * 2017-04-28 2020-03-04 Novartis AG Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
EP3409322A1 (en) 2017-06-01 2018-12-05 F. Hoffmann-La Roche AG Treatment method
PE20191846A1 (en) 2017-06-02 2019-12-31 Pfizer ANTIBODIES SPECIFIC TO FLT3 AND ITS USES
JP2020523383A (en) * 2017-06-14 2020-08-06 ダナ−ファーバー キャンサー インスティテュート,インコーポレイテッド B cell maturation antigen (BCMA)-directed nanoparticles
EP3642229A1 (en) 2017-06-21 2020-04-29 Gilead Sciences, Inc. Multispecific antibodies that target hiv gp120 and cd3
AU2018311503C1 (en) 2017-08-01 2023-11-30 Medimmune, Llc BCMA monoclonal antibody-drug conjugate
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
CN111107874A (en) * 2017-09-14 2020-05-05 葛兰素史密斯克莱知识产权发展有限公司 Combination therapy for cancer
EP3694878A1 (en) * 2017-09-14 2020-08-19 GlaxoSmithKline Intellectual Property Development Limited Combination treatment for cancer
EP3689908A4 (en) * 2017-09-29 2021-09-29 Mogam Institute for Biomedical Research Anti-bcma antibody having high affinity for bcma and pharmaceutical composition for treatment of cancer, comprising same
KR20200086278A (en) 2017-10-18 2020-07-16 노파르티스 아게 Compositions and methods for selective proteolysis
JP2021502979A (en) 2017-11-15 2021-02-04 ノバルティス アーゲー BCMA targeting chimeric antigen receptor, CD19 targeting chimeric antigen receptor and combination therapy
TW201925782A (en) 2017-11-30 2019-07-01 瑞士商諾華公司 BCMA-targeting chimeric antigen receptor, and uses thereof
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
AU2019215031A1 (en) 2018-01-31 2020-08-20 Novartis Ag Combination therapy using a chimeric antigen receptor
AU2019214183B2 (en) * 2018-02-01 2022-04-07 Innovent Biologics (Suzhou) Co., Ltd. Fully human anti-B cell maturation antigen (BCMA) single chain variable fragment, and application thereof
CN111936519A (en) 2018-02-01 2020-11-13 辉瑞公司 Antibodies specific for CD70 and uses thereof
MX2020008184A (en) 2018-02-01 2020-09-22 Pfizer Chimeric antigen receptors targeting cd70.
TWI829667B (en) 2018-02-09 2024-01-21 瑞士商赫孚孟拉羅股份公司 Antibodies binding to gprc5d
US20200399383A1 (en) 2018-02-13 2020-12-24 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
WO2019164891A1 (en) 2018-02-21 2019-08-29 Celgene Corporation Bcma-binding antibodies and uses thereof
CA3034795A1 (en) * 2018-02-27 2019-08-27 Pfizer Inc. Antibody purification
US11993658B2 (en) 2018-03-26 2024-05-28 Sutro Biopharma, Inc. Anti-BCMA antibodies and treatment methods
PE20210127A1 (en) 2018-05-23 2021-01-19 Pfizer CD3 SPECIFIC ANTIBODIES AND THEIR USES
EP3796912B1 (en) * 2018-05-23 2023-02-15 Celgene Corporation Antiproliferative compounds and bispecific antibody against bcma and cd3 for combined use
TWI803637B (en) 2018-05-23 2023-06-01 美商輝瑞大藥廠 Antibodies specific for gucy2c and uses thereof
JP7398396B2 (en) 2018-06-01 2023-12-14 ノバルティス アーゲー Binding molecules for BCMA and their uses
AU2019284911A1 (en) 2018-06-13 2020-12-17 Novartis Ag BCMA chimeric antigen receptors and uses thereof
CA3100187A1 (en) * 2018-06-26 2020-01-02 Abl Bio, Inc. Anti-bcma antibody and use thereof
US11845797B2 (en) 2018-07-03 2023-12-19 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
TWI838389B (en) 2018-07-19 2024-04-11 美商再生元醫藥公司 BISPECIFIC ANTI-BCMAxANTI-CD3 ANTIBODIES AND USES THEREOF
EP3844265A2 (en) 2018-08-31 2021-07-07 Novartis AG Methods of making chimeric antigen receptor-expressing cells
SG11202101825QA (en) 2018-08-31 2021-03-30 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
TW202029980A (en) * 2018-10-26 2020-08-16 美商免疫遺傳股份有限公司 Epcam antibodies, activatable antibodies, and immunoconjugates, and uses thereof
KR20210099574A (en) * 2018-12-01 2021-08-12 알로젠 테라퓨틱스 인코포레이티드 Chimeric antigen receptors targeting B-cell maturation antigens and methods of use thereof
EP3898682A1 (en) * 2018-12-21 2021-10-27 F. Hoffmann-La Roche AG Tumor-targeted agonistic cd28 antigen binding molecules
WO2020128893A1 (en) 2018-12-21 2020-06-25 Pfizer Inc. Combination treatments of cancer comprising a tlr agonist
SG11202107825WA (en) 2019-02-25 2021-09-29 Novartis Ag Mesoporous silica particles compositions for viral delivery
MX2021010281A (en) * 2019-02-26 2021-09-23 Sorrento Therapeutics Inc Antigen binding proteins that bind bcma.
MX2021010444A (en) 2019-03-01 2021-09-21 Allogene Therapeutics Inc Chimeric cytokine receptors bearing a pd-1 ectodomain.
SG11202109002XA (en) 2019-03-21 2021-09-29 Regeneron Pharma Combination of il-4/il-13 pathway inhibitors and plasma cell ablation for treating allergy
EP3942025A1 (en) 2019-03-21 2022-01-26 Novartis AG Car-t cell therapies with enhanced efficacy
US20220168389A1 (en) 2019-04-12 2022-06-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3959320A1 (en) 2019-04-24 2022-03-02 Novartis AG Compositions and methods for selective protein degradation
CN114286828A (en) 2019-06-24 2022-04-05 诺华股份有限公司 Dosing regimens and combination therapies for multispecific antibodies targeting B cell maturation antigens
US11339159B2 (en) 2019-07-17 2022-05-24 Pfizer Inc. Toll-like receptor agonists
US20220251228A1 (en) * 2019-07-30 2022-08-11 Shanghai Hansoh Biomedical Co., Ltd. Anti-bcma antibody, antigen-binding fragment thereof and medical use thereof
JP2022543551A (en) 2019-07-31 2022-10-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibody that binds to GPRC5D
PE20220394A1 (en) 2019-07-31 2022-03-18 Hoffmann La Roche ANTIBODIES THAT BIND TO GPRC5D
KR20220041915A (en) * 2019-08-06 2022-04-01 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Biopharmaceutical compositions and related methods
BR112022010206A2 (en) 2019-11-26 2022-11-29 Novartis Ag CHIMERIC ANTIGEN RECEPTORS AND USES THEREOF
JP2023505218A (en) * 2019-12-06 2023-02-08 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Methods of treating multiple myeloma with bispecific anti-BCMA x anti-CD3 antibodies
US20230039487A1 (en) * 2019-12-17 2023-02-09 Shenzhen Feipeng Biological Therapy Co. Ltd BCMA-binding antibody and use thereof
IL293926A (en) 2019-12-17 2022-08-01 Pfizer Antibodies specific for cd47, pd-l1, and uses thereof
TW202128176A (en) 2019-12-18 2021-08-01 美商輝瑞股份有限公司 Once daily cancer treatment regimen with a prmt5 inhibitor
KR20220012314A (en) * 2020-01-03 2022-02-03 살루브리스 (청두) 바이오테크 코., 리미티드 Antibodies that bind BCMA and uses thereof
CN111234020B (en) * 2020-01-23 2020-10-23 和铂医药(苏州)有限公司 BCMA binding protein and preparation method and application thereof
CN115397460A (en) 2020-02-27 2022-11-25 诺华股份有限公司 Methods of making cells expressing chimeric antigen receptors
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US20230095053A1 (en) * 2020-03-03 2023-03-30 Sutro Biopharma, Inc. Antibodies comprising site-specific glutamine tags, methods of their preparation and methods of their use
EP4118105A2 (en) 2020-03-09 2023-01-18 Pfizer Inc. Cd80-fc fusion protein and uses thereof
CN115279781A (en) * 2020-03-26 2022-11-01 上海翰森生物医药科技有限公司 Antibody drug conjugate and medical application thereof
CN111808191A (en) * 2020-05-11 2020-10-23 廊坊天光生物技术有限公司 Antibody pair for detecting VEGF content in serum and application thereof
BR112022022800A2 (en) * 2020-05-11 2022-12-13 Janssen Biotech Inc METHODS FOR TREATMENT OF MULTIPLE MYELOMA
BR112022021956A2 (en) 2020-05-13 2022-12-13 Pfizer METHODS, THERAPIES AND USES FOR THE TREATMENT OF CANCER
US20230332104A1 (en) 2020-06-11 2023-10-19 Novartis Ag Zbtb32 inhibitors and uses thereof
AU2021308586A1 (en) 2020-07-17 2023-03-02 Pfizer Inc. Therapeutic antibodies and their uses
AU2021329404A1 (en) 2020-08-21 2023-04-20 Novartis Ag Compositions and methods for in vivo generation of car expressing cells
AU2021339096A1 (en) 2020-09-14 2023-04-06 Pfizer Inc. Methods, therapies and uses for treating cancer
WO2022097090A1 (en) 2020-11-05 2022-05-12 Novartis Ag Dosing regimen for combination therapies with multispecific antibodies targeting b-cell maturation antigen and gamma secretase inhibitors
WO2022153161A1 (en) 2021-01-14 2022-07-21 Pfizer Inc. Treatment of cancer using a prmt5 inhibitor
KR20230157315A (en) 2021-01-28 2023-11-16 리제너론 파마슈티칼스 인코포레이티드 Compositions and methods for treating cytokine release syndrome
WO2022177676A1 (en) * 2021-02-16 2022-08-25 City Of Hope Anti-domain iv egfr antibodies and uses thereof
BR112023020832A2 (en) 2021-04-08 2023-12-19 Marengo Therapeutics Inc TCR-BINDED MULTIFUNCTIONAL MOLECULES AND THEIR USES
CN113358546A (en) * 2021-04-21 2021-09-07 贵州安康医学检验中心有限公司 Combined detection method for autoimmune peripheral neuropathy related antibody
AU2022267891A1 (en) 2021-04-27 2023-11-09 Novartis Ag Viral vector production system
JP2024519205A (en) 2021-04-30 2024-05-09 セルジーン コーポレーション Combination therapy using anti-BCMA antibody drug conjugates (ADCs) in combination with gamma secretase inhibitors (GSIs)
WO2023010068A2 (en) * 2021-07-28 2023-02-02 Cartesian Therapeutics, Inc. Multiprotein-engineered cells secreting a multispecific antibody
EP4386007A1 (en) 2021-08-12 2024-06-19 Shanghai TTM-BIO Technology Co., Ltd. Bispecific recombinant protein and use thereof
EP4388000A1 (en) 2021-08-20 2024-06-26 Novartis AG Methods of making chimeric antigen receptor?expressing cells
WO2023028612A2 (en) 2021-08-27 2023-03-02 Board Of Regents, The University Of Texas System Anti-tslpr (crlf2) antibodies
CN118043356A (en) * 2021-09-27 2024-05-14 Xencor股份有限公司 B Cell Maturation Antigen (BCMA) binding domain compositions
AR127484A1 (en) * 2021-10-29 2024-01-31 Lilly Co Eli COMPOUNDS AND METHODS TARGETTING INTERLEUKIN-34
WO2023166420A1 (en) 2022-03-03 2023-09-07 Pfizer Inc. Multispecific antibodies and uses thereof
WO2023201226A1 (en) 2022-04-11 2023-10-19 Regeneron Pharmaceuticals, Inc. Compositions and methods for universal tumor cell killing
WO2023218320A1 (en) 2022-05-11 2023-11-16 Pfizer Inc. Anti-lymphotoxin beta receptor antibodies and methods of use thereof
WO2023242769A1 (en) 2022-06-17 2023-12-21 Pfizer Inc. Il-12 variants, anti-pd1 antibodies, fusion proteins, and uses thereof
WO2024003773A1 (en) 2022-07-01 2024-01-04 Pfizer Inc. 2,7-naphthyridine compounds as mastl inhibitors
WO2024009191A1 (en) 2022-07-05 2024-01-11 Pfizer Inc. Pyrido[4,3-d]pyrimidine compounds
WO2024074977A1 (en) 2022-10-04 2024-04-11 Pfizer Inc. Substituted 1 h-pyrazolo-pyridine and-pyrimidine compounds
WO2024084364A1 (en) 2022-10-18 2024-04-25 Pfizer Inc. Compounds for the treatment of cancer
WO2024089639A1 (en) 2022-10-26 2024-05-02 Novartis Ag Lentiviral formulations
WO2024088987A1 (en) 2022-10-26 2024-05-02 F. Hoffmann-La Roche Ag Combination therapy for the treatment of cancer
WO2024105563A1 (en) 2022-11-16 2024-05-23 Pfizer Inc. Substituted bicyclic pyridone derivatives
WO2024121777A1 (en) 2022-12-09 2024-06-13 Pfizer Inc. Cd47 blocking agent and anti-bcma / anti-cd3 bispecific antibody combination therapy

Family Cites Families (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901464A (en) 1943-01-23 1945-07-27 Boehringer & Soehne Gmbh Process for obtaining vanillin
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
FR2413974A1 (en) 1978-01-06 1979-08-03 David Bernard DRYER FOR SCREEN-PRINTED SHEETS
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4754065A (en) 1984-12-18 1988-06-28 Cetus Corporation Precursor to nucleic acid probe
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
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
US4777127A (en) 1985-09-30 1988-10-11 Labsystems Oy Human retrovirus-related products and methods of diagnosing and treating conditions associated with said retrovirus
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
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
GB8702816D0 (en) 1987-02-07 1987-03-11 Al Sumidaie A M K Obtaining retrovirus-containing fraction
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
US5422120A (en) 1988-05-30 1995-06-06 Depotech Corporation Heterovesicular liposomes
AP129A (en) 1988-06-03 1991-04-17 Smithkline Biologicals S A Expression of retrovirus gag protein eukaryotic cells
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
EP0454781B1 (en) 1989-01-23 1998-12-16 Chiron Corporation Recombinant cells for therapies of infection and hyperproliferative disorders and preparation thereof
US6673776B1 (en) 1989-03-21 2004-01-06 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
DE69032284T2 (en) 1989-03-21 1998-10-08 Vical, Inc., San Diego, Calif. EXPRESSION OF EXOGENOUS POLYNUCLEOTIDE SEQUENCES IN VERTEBLE
EP0739904A1 (en) 1989-06-29 1996-10-30 Medarex, Inc. Bispecific reagents for aids therapy
WO1991002805A2 (en) 1989-08-18 1991-03-07 Viagene, Inc. Recombinant retroviruses delivering vector constructs to target cells
US5585362A (en) 1989-08-22 1996-12-17 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
ZA911974B (en) 1990-03-21 1994-08-22 Res Dev Foundation Heterovesicular liposomes
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
WO1992000373A1 (en) 1990-06-29 1992-01-09 Biosource Genetics Corporation Melanin production by transformed microorganisms
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
KR100272077B1 (en) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
CA2405246A1 (en) 1990-12-03 1992-06-11 Genentech, Inc. Enrichment method for variant proteins with alterred binding properties
US5278299A (en) 1991-03-18 1994-01-11 Scripps Clinic And Research Foundation Method and composition for synthesizing sialylated glycosyl compounds
EP0586505A1 (en) 1991-05-14 1994-03-16 Repligen Corporation Heteroconjugate antibodies for treatment of hiv infection
EP0519596B1 (en) 1991-05-17 2005-02-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
CA2103059C (en) 1991-06-14 2005-03-22 Paul J. Carter Method for making humanized antibodies
GB9115364D0 (en) 1991-07-16 1991-08-28 Wellcome Found Antibody
ES2197145T3 (en) 1991-08-20 2004-01-01 The Government Of The Usa As Represented By The Secretary Of The Deptm. Of Health And Human Services TRANSFER OF GENES MEDIATED BY ADENOVIRUS TO GASTROINTESTINAL.
AU669124B2 (en) 1991-09-18 1996-05-30 Kyowa Hakko Kirin Co., Ltd. Process for producing humanized chimera antibody
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993010218A1 (en) 1991-11-14 1993-05-27 The United States Government As Represented By The Secretary Of The Department Of Health And Human Services Vectors including foreign genes and negative selective markers
AU3144193A (en) 1991-11-21 1993-06-15 Board Of Trustees Of The Leland Stanford Junior University Controlling degradation of glycoprotein oligosaccharides by extracellular glycosisases
GB9125623D0 (en) 1991-12-02 1992-01-29 Dynal As Cell modification
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
FR2688514A1 (en) 1992-03-16 1993-09-17 Centre Nat Rech Scient Defective recombinant adenoviruses expressing cytokines and antitumour drugs containing them
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1993025234A1 (en) 1992-06-08 1993-12-23 The Regents Of The University Of California Methods and compositions for targeting specific tissue
WO1993025698A1 (en) 1992-06-10 1993-12-23 The United States Government As Represented By The Vector particles resistant to inactivation by human serum
GB2269175A (en) 1992-07-31 1994-02-02 Imperial College Retroviral vectors
ATE149570T1 (en) 1992-08-17 1997-03-15 Genentech Inc BISPECIFIC IMMUNOADHESINS
US5837821A (en) 1992-11-04 1998-11-17 City Of Hope Antibody construct
US6210671B1 (en) 1992-12-01 2001-04-03 Protein Design Labs, Inc. Humanized antibodies reactive with L-selectin
CA2592997A1 (en) 1992-12-03 1994-06-09 Genzyme Corporation Pseudo-adenovirus vectors
US5981568A (en) 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
JP3545403B2 (en) 1993-04-22 2004-07-21 スカイファルマ インコーポレイテッド Cyclodextrin liposomes encapsulating pharmaceutical compounds and methods of use
US6180377B1 (en) 1993-06-16 2001-01-30 Celltech Therapeutics Limited Humanized antibodies
EP0705344B8 (en) 1993-06-24 2006-05-10 Advec Inc. Adenovirus vectors for gene therapy
EP0694070B1 (en) 1993-09-15 2002-04-10 Chiron Corporation Recombinant alphavirus vectors
US6015686A (en) 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
BR9407956A (en) 1993-10-25 1996-11-26 Canji Inc Pharmaceutical composition vector of recombinant adenovirus expression and kit to reduce tumor cell proliferation
NZ276305A (en) 1993-11-16 1997-10-24 Depotech Corp Controlled release vesicle compositions
US6436908B1 (en) 1995-05-30 2002-08-20 Duke University Use of exogenous β-adrenergic receptor and β-adrenergic receptor kinase gene constructs to enhance myocardial function
JP4303315B2 (en) 1994-05-09 2009-07-29 オックスフォード バイオメディカ(ユーケー)リミテッド Non-crossing retroviral vector
WO1996017072A2 (en) 1994-11-30 1996-06-06 Chiron Viagene, Inc. Recombinant alphavirus vectors
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
US6265150B1 (en) 1995-06-07 2001-07-24 Becton Dickinson & Company Phage antibodies
AU2998597A (en) 1996-05-06 1997-11-26 Chiron Corporation Crossless retroviral vectors
AU9692198A (en) 1997-10-10 1999-05-03 Kevin J. Donahue Gene delivery compositions and methods
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2000053211A2 (en) 1999-03-09 2000-09-14 University Of Southern California Method of promoting myocyte proliferation and myocardial tissue repair
MXPA02001665A (en) * 1999-08-17 2003-07-14 Biogen Inc Baff receptor (bcma), an immunoregulatory agent.
US6849425B1 (en) 1999-10-14 2005-02-01 Ixsys, Inc. Methods of optimizing antibody variable region binding affinity
US20080267965A1 (en) 2002-02-21 2008-10-30 Kalled Susan L Use of Bcma as an Immunoregulatory Agent
AU2003232456B2 (en) * 2002-05-30 2009-06-04 Macrogenics, Inc. CD16A binding proteins and use for the treatment of immune disorders
KR101250818B1 (en) 2002-12-24 2013-04-15 리나트 뉴로사이언스 코프. Anti-ngf antibodies and methods using same
CA2604238C (en) 2005-04-15 2015-07-07 Neogenix Oncology, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
ES2593583T3 (en) * 2009-03-10 2016-12-09 Biogen Ma Inc. Anti-BCMA antibodies
AU2010317063B2 (en) 2009-11-04 2014-10-23 Dsm Ip Assets B.V. Talaromyces transformants
EP2569337A1 (en) 2010-05-14 2013-03-20 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
US9676871B2 (en) 2010-11-05 2017-06-13 Pfizer Inc. Engineered polypeptide conjugates and methods for making thereof using transglutaminase
US20130273055A1 (en) * 2010-11-16 2013-10-17 Eric Borges Agents and methods for treating diseases that correlate with bcma expression
US20130101599A1 (en) * 2011-04-21 2013-04-25 Boehringer Ingelheim International Gmbh Bcma-based stratification and therapy for multiple myeloma patients
DK3415531T3 (en) * 2011-05-27 2023-09-18 Glaxo Group Ltd BCMA (CD269/TNFRSF17)-BINDING PROTEINS
TWI679212B (en) 2011-11-15 2019-12-11 美商安進股份有限公司 Binding molecules for e3 of bcma and cd3
BR112014012609A2 (en) 2011-11-17 2020-10-20 Pfizer Inc cytotoxic and conjugated antibody drug peptides thereof
EP2836239A1 (en) 2012-04-11 2015-02-18 The United States of America, as Represented By the Secretary, Department of Health and Human Services Chimeric antigen receptors targeting b-cell maturation antigen
CA2870545A1 (en) 2012-04-20 2013-10-24 Emergent Product Development Seattle, Llc Cd3 binding polypeptides
EP2914628A1 (en) 2012-11-01 2015-09-09 Max-Delbrück-Centrum für Molekulare Medizin An antibody that binds cd269 (bcma) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
TW201425336A (en) * 2012-12-07 2014-07-01 Amgen Inc BCMA antigen binding proteins
US9243058B2 (en) 2012-12-07 2016-01-26 Amgen, Inc. BCMA antigen binding proteins
WO2014122144A1 (en) * 2013-02-05 2014-08-14 Engmab Ag BISPECIFIC ANTIBODIES AGAINST CD3ε AND BCMA
EP2762497A1 (en) * 2013-02-05 2014-08-06 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
AR095374A1 (en) 2013-03-15 2015-10-14 Amgen Res (Munich) Gmbh UNION MOLECULES FOR BCMA AND CD3
WO2015015448A2 (en) 2013-07-31 2015-02-05 Rinat Neuroscience Corp. Engineered polypeptide conjugates
GB201317928D0 (en) 2013-10-10 2013-11-27 Ucl Business Plc Molecule
ES2811923T3 (en) * 2013-12-17 2021-03-15 Genentech Inc Anti-CD3 antibodies and methods of use
JP6755805B2 (en) 2014-04-30 2020-09-16 マックス−デルブリュック−ツェントルム フューア モレキュラーレ メディツィン イン デア ヘルムホルツ−ゲマインシャフト Humanized antibody against CD269 (BCMA)
EP3023437A1 (en) 2014-11-20 2016-05-25 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
EP3029068A1 (en) 2014-12-03 2016-06-08 EngMab AG Bispecific antibodies against CD3epsilon and BCMA for use in the treatment of diseases
BR112017019914A2 (en) 2015-04-13 2018-06-19 Pfizer b-maturation antigen-directed chimeric antigen receptors

Also Published As

Publication number Publication date
IL290496A (en) 2022-04-01
WO2016166629A1 (en) 2016-10-20
MX2017013297A (en) 2018-06-19
AU2016248946A1 (en) 2017-09-21
IL290488B2 (en) 2024-07-01
US10793635B2 (en) 2020-10-06
PE20221580A1 (en) 2022-10-06
ZA201807153B (en) 2022-08-31
PH12017501732A1 (en) 2018-04-02
IL310723A (en) 2024-04-01
PE20180046A1 (en) 2018-01-15
CO2017010495A2 (en) 2018-01-05
CA3219680A1 (en) 2016-10-13
FIC20240014I1 (en) 2024-05-21
KR20170131699A (en) 2017-11-29
TW201811833A (en) 2018-04-01
CA2925329A1 (en) 2016-10-13
SI3283106T1 (en) 2022-04-29
MX2022001831A (en) 2022-03-17
CN108136002B (en) 2022-05-31
HK1255385A1 (en) 2019-08-16
CN114773476A (en) 2022-07-22
US20210188991A1 (en) 2021-06-24
EP3283106B1 (en) 2021-12-22
TWI703157B (en) 2020-09-01
CA3219684A1 (en) 2016-10-13
KR102405099B1 (en) 2022-06-07
AU2016248946B2 (en) 2022-03-10
US11155630B2 (en) 2021-10-26
US20160297885A1 (en) 2016-10-13
EP3283106A1 (en) 2018-02-21
ES2905315T3 (en) 2022-04-07
KR20200053641A (en) 2020-05-18
IL254219B (en) 2022-03-01
IL290488A (en) 2022-04-01
JP2021169486A (en) 2021-10-28
IL254219A0 (en) 2017-10-31
US9969809B2 (en) 2018-05-15
SA517390140B1 (en) 2021-11-24
CN108136002A (en) 2018-06-08
BR112017019785B1 (en) 2022-11-16
JP6913025B2 (en) 2021-08-04
KR102110557B1 (en) 2020-05-15
HUE057285T2 (en) 2022-04-28
EP4234581A3 (en) 2023-10-04
SG10201912823PA (en) 2020-02-27
CN114874331A (en) 2022-08-09
RU2017133826A (en) 2019-05-13
JP2018516068A (en) 2018-06-21
US10040860B2 (en) 2018-08-07
RU2722381C2 (en) 2020-05-29
SG11201707192XA (en) 2017-10-30
CA2925329C (en) 2024-01-02
AU2022203601A1 (en) 2022-06-16
AU2022203620A1 (en) 2022-06-16
PT3283106T (en) 2022-02-02
MX2022001830A (en) 2022-03-17
PL3283106T3 (en) 2022-05-02
US11814435B2 (en) 2023-11-14
DK3283106T3 (en) 2022-01-10
IL290488B1 (en) 2024-03-01
TW201706311A (en) 2017-02-16
PE20221262A1 (en) 2022-08-16
JP2023109898A (en) 2023-08-08
SG10201912819XA (en) 2020-02-27
MY196625A (en) 2023-04-23
TWI703159B (en) 2020-09-01
CA3219665A1 (en) 2016-10-13
EP4234581A2 (en) 2023-08-30
KR20200053642A (en) 2020-05-18
BR112017019785A2 (en) 2018-05-22
EP3988117A1 (en) 2022-04-27
US20210054087A1 (en) 2021-02-25
TW202115120A (en) 2021-04-16
US20180171018A1 (en) 2018-06-21
TWI787645B (en) 2022-12-21
KR102405104B1 (en) 2022-06-07
RU2017133826A3 (en) 2019-05-13
US20180298108A1 (en) 2018-10-18

Similar Documents

Publication Publication Date Title
US11155630B2 (en) Therapeutic antibodies and their uses
US20190194335A1 (en) Antibodies specific for epidermal growth factor receptor variant iii and their uses

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION